Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Rep ; 13(1): 22000, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-38081972

RESUMEN

The histone deacetylase sirtuin 6 (SIRT6) has been endowed with anti-cancer capabilities in many tumor types. Here, we investigate the impact of SIRT6-overexpression (SIRT6-OE) in Delta16HER2 mice, which are a bona fide model of HER2-positive breast cancer. After an initial delay in the tumor onset, SIRT6-OE induces a more aggressive phenotype of Delta16HER2 tumors promoting the formation of higher number of tumor foci and metastases than controls. This phenotype of SIRT6-OE tumors is associated with cancer stem cell (CSC)-like features and tumor dormancy, and low senescence and oxidative DNA damage. Accordingly, a sub-set of HER2-positive breast cancer patients with concurrent SIRT6-OE has a significant poorer relapse-free survival (RFS) probability than patients with low expression of SIRT6. ChIP-seq, RNA-seq and RT-PCR experiments indicate that SIRT6-OE represses the expression of the T-box transcription factor 3 (Tbx3) by deacetylation of H3K9ac. Accordingly, loss-of-function mutations of TBX3 or low TBX3 expression levels are predictive of poor prognosis in HER2-positive breast cancer patients. Our work indicates that high levels of SIRT6 are indicative of poor prognosis and high risk of metastasis in HER2-positive breast cancer and suggests further investigation of TBX3 as a downstream target of SIRT6 and co-marker of poor-prognosis. Our results point to a breast cancer subtype-specific effect of SIRT6 and warrant future studies dissecting the mechanisms of SIRT6 regulation in different breast cancer subtypes.


Asunto(s)
Neoplasias de la Mama , Sirtuinas , Humanos , Animales , Ratones , Femenino , Neoplasias de la Mama/patología , Recurrencia Local de Neoplasia , Sirtuinas/metabolismo , Enfermedad Crónica
2.
Nat Commun ; 12(1): 5068, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34417460

RESUMEN

p53 regulates several signaling pathways to maintain the metabolic homeostasis of cells and modulates the cellular response to stress. Deficiency or excess of nutrients causes cellular metabolic stress, and we hypothesized that p53 could be linked to glucose maintenance. We show here that upon starvation hepatic p53 is stabilized by O-GlcNAcylation and plays an essential role in the physiological regulation of glucose homeostasis. More specifically, p53 binds to PCK1 promoter and regulates its transcriptional activation, thereby controlling hepatic glucose production. Mice lacking p53 in the liver show a reduced gluconeogenic response during calorie restriction. Glucagon, adrenaline and glucocorticoids augment protein levels of p53, and administration of these hormones to p53 deficient human hepatocytes and to liver-specific p53 deficient mice fails to increase glucose levels. Moreover, insulin decreases p53 levels, and over-expression of p53 impairs insulin sensitivity. Finally, protein levels of p53, as well as genes responsible of O-GlcNAcylation are elevated in the liver of type 2 diabetic patients and positively correlate with glucose and HOMA-IR. Overall these results indicate that the O-GlcNAcylation of p53 plays an unsuspected key role regulating in vivo glucose homeostasis.


Asunto(s)
Acetilglucosamina/metabolismo , Glucosa/metabolismo , Hígado/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Secuencia de Bases , Restricción Calórica , Línea Celular , Colforsina/farmacología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Epinefrina/metabolismo , Glucagón/metabolismo , Glucocorticoides/metabolismo , Gluconeogénesis/efectos de los fármacos , Glicosilación , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hidrocortisona/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Resistencia a la Insulina , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/complicaciones , Obesidad/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Ácido Pirúvico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética
3.
Nat Commun ; 11(1): 5808, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33199701

RESUMEN

Skeletal muscle promotes metabolic balance by regulating glucose uptake and the stimulation of multiple interorgan crosstalk. We show here that the catalytic activity of Vav2, a Rho GTPase activator, modulates the signaling output of the IGF1- and insulin-stimulated phosphatidylinositol 3-kinase pathway in that tissue. Consistent with this, mice bearing a Vav2 protein with decreased catalytic activity exhibit reduced muscle mass, lack of proper insulin responsiveness and, at much later times, a metabolic syndrome-like condition. Conversely, mice expressing a catalytically hyperactive Vav2 develop muscle hypertrophy and increased insulin responsiveness. Of note, while hypoactive Vav2 predisposes to, hyperactive Vav2 protects against high fat diet-induced metabolic imbalance. These data unveil a regulatory layer affecting the signaling output of insulin family factors in muscle.


Asunto(s)
Biocatálisis , Homeostasis , Metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Biocatálisis/efectos de los fármacos , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Genotipo , Glucosa/farmacología , Homeostasis/efectos de los fármacos , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Músculo Esquelético/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo
4.
Cell Rep ; 30(11): 3851-3863.e6, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32187554

RESUMEN

Cancer therapy is limited, in part, by lack of specificity. Thus, identifying molecules that are selectively expressed by, and relevant for, cancer cells is of paramount medical importance. Here, we show that peptidyl-prolyl-cis-trans-isomerase (PPIase) FK506-binding protein 10 (FKBP10)-positive cells are present in cancer lesions but absent in the healthy parenchyma of human lung. FKBP10 expression negatively correlates with survival of lung cancer patients, and its downregulation causes a dramatic diminution of lung tumor burden in mice. Mechanistically, our results from gain- and loss-of-function assays show that FKBP10 boosts cancer growth and stemness via its PPIase activity. Also, FKBP10 interacts with ribosomes, and its downregulation leads to reduction of translation elongation at the beginning of open reading frames (ORFs), particularly upon insertion of proline residues. Thus, our data unveil FKBP10 as a cancer-selective molecule with a key role in translational reprogramming, stem-like traits, and growth of lung cancer.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Biosíntesis de Proteínas , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Isomerasa de Peptidilprolil/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ribosomas/metabolismo
5.
Nat Commun ; 8(1): 1820, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29180649

RESUMEN

Obesity-induced inflammation engenders insulin resistance and type 2 diabetes mellitus (T2DM) but the inflammatory effectors linking obesity to insulin resistance are incompletely understood. Here, we show that hepatic expression of Protein Tyrosine Phosphatase Receptor Gamma (PTPR-γ) is stimulated by inflammation in obese/T2DM mice and positively correlates with indices of inflammation and insulin resistance in humans. NF-κB binds to the promoter of Ptprg and is required for inflammation-induced PTPR-γ expression. PTPR-γ loss-of-function lowers glycemia and insulinemia by enhancing insulin-stimulated suppression of endogenous glucose production. These phenotypes are rescued by re-expression of Ptprg only in liver of mice lacking Ptprg globally. Hepatic PTPR-γ overexpression that mimics levels found in obesity is sufficient to cause severe hepatic and systemic insulin resistance. We propose hepatic PTPR-γ as a link between obesity-induced inflammation and insulin resistance and as potential target for treatment of T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Obesidad/metabolismo , Proteínas Tirosina Fosfatasas Similares a Receptores/metabolismo , Adulto , Anciano , Animales , Glucemia , Línea Celular , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Células Hep G2 , Humanos , Inflamación/metabolismo , Insulina/sangre , Interleucina-6/metabolismo , Metabolismo de los Lípidos , Lipopolisacáridos/efectos adversos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Persona de Mediana Edad , Modelos Animales , FN-kappa B/metabolismo , Obesidad/sangre , Obesidad/complicaciones , Proteínas Tirosina Fosfatasas/metabolismo , ARN Mensajero/biosíntesis , Proteínas Tirosina Fosfatasas Similares a Receptores/genética , Sirtuina 1/metabolismo
6.
Cell Rep ; 18(8): 1858-1868, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28228253

RESUMEN

Cancer stem cells (CSCs) have high tumorigenic capacity. Here, we show that stem-like traits of specific human cancer cells are reduced by overexpression of the histone deacetylase sirtuin 6 (SIRT6). SIRT6-sensitive cancer cells bear mutations that activate phosphatidylinositol-3-kinase (PI3K) signaling, and overexpression of SIRT6 reduces growth, progression, and grade of breast cancer in a mouse model with PI3K activation. Tumor metabolomic and transcriptomic analyses reveal that SIRT6 overexpression dampens PI3K signaling and stem-like characteristics and causes metabolic rearrangements in this cancer model. Ablation of a PI3K activating mutation in otherwise isogenic cancer cells is sufficient to convert SIRT6-sensitive into SIRT6-insensitive cells. SIRT6 overexpression suppresses PI3K signaling at the transcriptional level and antagonizes tumor sphere formation independent of its histone deacetylase activity. Our data identify SIRT6 as a putative molecular target that hinders stemness of tumors with PI3K activation.


Asunto(s)
Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Sirtuinas/metabolismo , Acetilación , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación/fisiología , Transducción de Señal/fisiología , Transcripción Genética/fisiología
7.
Cell Metab ; 21(1): 117-25, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25533479

RESUMEN

Dietary effects on tumor biology can be exploited to unravel cancer vulnerabilities. Here, we present surprising evidence for anti-proliferative action of high-calorie-diet (HCD) feeding on KRAS-driven lung tumors. Tumors of mice that commenced HCD feeding before tumor onset displayed defective unfolded protein response (UPR) and unresolved endoplasmic reticulum (ER) stress. Unresolved ER stress and reduced proliferation are reversed by chemical chaperone treatment. Whole-genome transcriptional analyses revealed FKBP10 as one of the most downregulated chaperones in tumors of the HCD-pre-tumor-onset group. FKBP10 downregulation dampens tumor growth in vitro and in vivo. Providing translational value to these results, we report that FKBP10 is expressed in human KRAS-positive and -negative lung cancers, but not in healthy parenchyma. Collectively, our data shed light on an unexpected anti-tumor action of HCD imposed before tumor onset and identify FKBP10 as a putative therapeutic target to selectively hinder lung cancer.


Asunto(s)
Dieta , Neoplasias Pulmonares/patología , Fenilbutiratos/toxicidad , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Regulación hacia Abajo , Doxiciclina/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones SCID , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Trasplante Heterólogo , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteínas ras/genética
8.
PLoS One ; 7(10): e48643, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23119079

RESUMEN

Clinical research shows an association between polycystic ovary syndrome (PCOS) and chronic inflammation, a pathological state thought to contribute to insulin resistance. The underlying pathways, however, have not been defined. The purpose of this study was to characterize the inflammatory state of a novel mouse model of PCOS. Female mice lacking leptin and insulin receptors in pro-opiomelanocortin neurons (IR/LepR(POMC) mice) and littermate controls were evaluated for estrous cyclicity, ovarian and adipose tissue morphology, and body composition by QMR and CT scan. Tissue-specific macrophage infiltration and cytokine mRNA expression were measured, as well as circulating cytokine levels. Finally, glucose regulation during pregnancy was evaluated as a measure of risk for diabetes development. Forty-five percent of IR/LepR(POMC) mice showed reduced or absent ovulation. IR/LepR(POMC) mice also had increased fat mass and adipocyte hypertrophy. These traits accompanied elevations in macrophage accumulation and inflammatory cytokine production in perigonadal adipose tissue, liver, and ovary. These mice also exhibited gestational hyperglycemia as predicted. This report is the first to show the presence of inflammation in IR/LepR(POMC) mice, which develop a PCOS-like phenotype. Thus, IR/LepR(POMC) mice may serve as a new mouse model to clarify the involvement of adipose and liver tissue in the pathogenesis and etiology of PCOS, allowing more targeted research on the development of PCOS and potential therapeutic interventions.


Asunto(s)
Adipocitos/metabolismo , Modelos Animales de Enfermedad , Inflamación/genética , Síndrome del Ovario Poliquístico/genética , Adipocitos/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Glucemia/metabolismo , Femenino , Expresión Génica , Humanos , Hipertrofia , Inflamación/metabolismo , Interleucina-1beta/sangre , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/sangre , Interleucina-6/genética , Interleucina-6/metabolismo , Hígado/metabolismo , Hígado/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Ovario/metabolismo , Ovario/patología , Ovulación/genética , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/metabolismo , Embarazo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tomografía Computarizada por Rayos X
9.
J Clin Invest ; 121(1): 355-68, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21183787

RESUMEN

Studies in humans and rodents indicate that a minimum amount of stored energy is required for normal pubertal development. The adipocyte-derived hormone leptin is a key metabolic signal to the neuroendocrine reproductive axis. Humans and mice lacking leptin or the leptin receptor (LepR) (ob/ob and db/db mice, respectively) are infertile and fail to enter puberty. Leptin administration to leptin-deficient subjects and ob/ob mice induces puberty and restores fertility, but the exact site or sites of leptin action are unclear. Here, we found that genetic deletion of LepR selectively from hypothalamic Kiss1 neurons in mice had no effect on puberty or fertility, indicating that direct leptin signaling in Kiss1 neurons is not required for these processes. However, bilateral lesions of the ventral premammillary nucleus (PMV) of ob/ob mice blunted the ability of exogenous leptin to induce sexual maturation. Moreover, unilateral reexpression of endogenous LepR in PMV neurons was sufficient to induce puberty and improve fertility in female LepR-null mice. This LepR reexpression also normalized the increased hypothalamic GnRH content characteristic of leptin-signaling deficiency. These data suggest that the PMV is a key site for leptin's permissive action at the onset of puberty and support the hypothesis that the multiple actions of leptin to control metabolism and reproduction are anatomically dissociated.


Asunto(s)
Hipotálamo/metabolismo , Leptina/metabolismo , Proteínas/metabolismo , Maduración Sexual/fisiología , Animales , Secuencia de Bases , Femenino , Fertilidad/genética , Fertilidad/fisiología , Expresión Génica , Humanos , Kisspeptinas , Leptina/deficiencia , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Neuronas/metabolismo , Embarazo , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Maduración Sexual/genética , Transducción de Señal
10.
Endocrinology ; 150(12): 5326-33, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19819963

RESUMEN

Resveratrol is a natural polyphenolic compound that activates nicotinamide adenosine dinucleotide-dependent deacetylase SIRT1. Resveratrol has recently been shown to exert potent antidiabetic actions when orally delivered to animal models of type 2 diabetes. However, the tissue(s) mediating these beneficial effects is unknown. Because SIRT1 is expressed in central nervous system (CNS) neurons known to control glucose and insulin homeostasis, we hypothesized that resveratrol antidiabetic effects are mediated by the brain. Here, we report that long-term intracerebroventricular infusion of resveratrol normalizes hyperglycemia and greatly improves hyperinsulinemia in diet-induced obese and diabetic mice. It is noteworthy that these effects are independent of changes in body weight, food intake, and circulating leptin levels. In addition, CNS resveratrol delivery improves hypothalamic nuclear factor-kappaB inflammatory signaling by reducing acetylated-RelA/p65 and total RelA/p65 protein contents, and inhibitor of nuclear factor-kappaB alpha and IkappaB kinase beta mRNA levels. Furthermore, this treatment leads to reduced hepatic phosphoenolpyruvate carboxykinase 1 mRNA and protein levels and ameliorates pyruvate-induced hyperglycemia in this mouse model of type 2 diabetes. Collectively, our results unveiled a previously unrecognized key role for the CNS in mediating the antidiabetic actions of resveratrol.


Asunto(s)
Encéfalo/efectos de los fármacos , Diabetes Mellitus Tipo 2/prevención & control , Sirtuina 1/metabolismo , Estilbenos/farmacología , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Western Blotting , Peso Corporal/efectos de los fármacos , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/etiología , Grasas de la Dieta , Ingestión de Alimentos/efectos de los fármacos , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Hiperglucemia/sangre , Hiperglucemia/etiología , Hiperglucemia/prevención & control , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Resveratrol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirtuina 1/genética , Estilbenos/administración & dosificación
11.
Nat Clin Pract Endocrinol Metab ; 5(3): 160-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19229236

RESUMEN

Individuals who live in industrialized countries often eat a calorie-rich diet and perform little physical activity. These habits are thought to be critical contributors to the rapidly rising incidence of obesity, a condition that affects hundreds of millions of people worldwide. High-calorie intake alters metabolic-sensing pathways in central nervous system neurons, and these changes have pathogenic roles in the development of obesity. This review aims to summarize our current knowledge about the neuronal populations (the central melanocortin system in particular) and transcriptional regulators, including STAT3 and FOXO1, that are involved in the maintenance of normal body weight. We describe the interactions between these transcriptional factors and their target genes, which encode the main appetite-regulating neuropeptides (agouti-related peptide and alpha-melanocyte-stimulating hormone). We discuss the transcriptional co-activator PGC-1-alpha and the supposed metabolic-sensor protein SIRT1, and their potential roles as targets for novel antiobesity medications.


Asunto(s)
Apetito/fisiología , Peso Corporal/fisiología , Factores de Transcripción/fisiología , Apetito/genética , Peso Corporal/genética , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/fisiología , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/fisiología , Modelos Biológicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/fisiología , Sirtuina 1 , Sirtuinas/genética , Sirtuinas/metabolismo , Sirtuinas/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
J Neurosci ; 28(40): 9989-96, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18829956

RESUMEN

SIRT1 is a nicotinamide adenosine dinucleotide-dependent deacetylase that orchestrates key metabolic adaptations to nutrient deprivation in peripheral tissues. SIRT1 is induced also in the brain by reduced energy intake. However, very little is known about SIRT1 distribution and the biochemical phenotypes of SIRT1-expressing cells in the neuraxis. Unknown are also the brain sites in which SIRT1 is regulated by energy availability and whether these regulations are altered in a genetic model of obesity. To address these issues, we performed in situ hybridization histochemistry analyses and found that Sirt1 mRNA is highly expressed in metabolically relevant sites. These include, but are not limited to, the hypothalamic arcuate, ventromedial, dorsomedial, and paraventricular nuclei and the area postrema and the nucleus of the solitary tract in the hindbrain. Of note, our single-cell reverse transcription-PCR analyses revealed that Sirt1 mRNA is expressed in pro-opiomelanocortin neurons that are critical for normal body weight and glucose homeostasis. We also found that SIRT1 protein levels are restrictedly increased in the hypothalamus in the fasted brain. Of note, we found that this hypothalamic-specific, fasting-induced SIRT1 regulation is altered in leptin-deficient, obese mice. Collectively, our findings establish the distribution of Sirt1 mRNA throughout the neuraxis and suggest a previously unrecognized role of brain SIRT1 in regulating energy homeostasis.


Asunto(s)
Química Encefálica/fisiología , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Metabolismo Energético/fisiología , Sirtuinas/metabolismo , Animales , Encéfalo/fisiología , Homeostasis/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Sirtuina 1 , Sirtuinas/fisiología
13.
Endocrinology ; 149(4): 1773-85, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18162515

RESUMEN

Two known types of leptin-responsive neurons reside within the arcuate nucleus: the agouti gene-related peptide (AgRP)/neuropeptide Y (NPY) neuron and the proopiomelanocortin (POMC) neuron. By deleting the leptin receptor gene (Lepr) specifically in AgRP/NPY and/or POMC neurons of mice, we examined the several and combined contributions of these neurons to leptin action. Body weight and adiposity were increased by Lepr deletion from AgRP and POMC neurons individually, and simultaneous deletion in both neurons (A+P LEPR-KO mice) further increased these measures. Young (periweaning) A+P LEPR-KO mice exhibit hyperphagia and decreased energy expenditure, with increased weight gain, oxidative sparing of triglycerides, and increased fat accumulation. Interestingly, however, many of these abnormalities were attenuated in adult animals, and high doses of leptin partially suppress food intake in the A+P LEPR-KO mice. Although mildly hyperinsulinemic, the A+P LEPR-KO mice displayed normal glucose tolerance and fertility. Thus, AgRP/NPY and POMC neurons each play mandatory roles in aspects of leptin-regulated energy homeostasis, high leptin levels in adult mice mitigate the importance of leptin-responsiveness in these neurons for components of energy balance, suggesting the presence of other leptin-regulated pathways that partially compensate for the lack of leptin action on the POMC and AgRP/NPY neurons.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Ingestión de Alimentos , Metabolismo Energético , Proopiomelanocortina/fisiología , Receptores de Leptina/fisiología , Animales , Composición Corporal , Fertilidad , Hiperinsulinismo/etiología , Hiperfagia , Lactancia , Masculino , Ratones , Neuropéptido Y/fisiología
14.
Nature ; 449(7159): 228-32, 2007 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-17728716

RESUMEN

A subset of neurons in the brain, known as 'glucose-excited' neurons, depolarize and increase their firing rate in response to increases in extracellular glucose. Similar to insulin secretion by pancreatic beta-cells, glucose excitation of neurons is driven by ATP-mediated closure of ATP-sensitive potassium (K(ATP)) channels. Although beta-cell-like glucose sensing in neurons is well established, its physiological relevance and contribution to disease states such as type 2 diabetes remain unknown. To address these issues, we disrupted glucose sensing in glucose-excited pro-opiomelanocortin (POMC) neurons via transgenic expression of a mutant Kir6.2 subunit (encoded by the Kcnj11 gene) that prevents ATP-mediated closure of K(ATP) channels. Here we show that this genetic manipulation impaired the whole-body response to a systemic glucose load, demonstrating a role for glucose sensing by POMC neurons in the overall physiological control of blood glucose. We also found that glucose sensing by POMC neurons became defective in obese mice on a high-fat diet, suggesting that loss of glucose sensing by neurons has a role in the development of type 2 diabetes. The mechanism for obesity-induced loss of glucose sensing in POMC neurons involves uncoupling protein 2 (UCP2), a mitochondrial protein that impairs glucose-stimulated ATP production. UCP2 negatively regulates glucose sensing in POMC neurons. We found that genetic deletion of Ucp2 prevents obesity-induced loss of glucose sensing, and that acute pharmacological inhibition of UCP2 reverses loss of glucose sensing. We conclude that obesity-induced, UCP2-mediated loss of glucose sensing in glucose-excited neurons might have a pathogenic role in the development of type 2 diabetes.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Neuronas/metabolismo , Obesidad/fisiopatología , Proopiomelanocortina/metabolismo , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Humanos , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/genética , Canales Iónicos/metabolismo , Glicósidos Iridoides , Iridoides/farmacología , Ratones , Ratones Obesos , Ratones Transgénicos , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Obesidad/inducido químicamente , Obesidad/metabolismo , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , Proteína Desacopladora 2
15.
Cell Metab ; 4(6): 453-64, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17141629

RESUMEN

PGC-1beta is a transcriptional coactivator that potently stimulates mitochondrial biogenesis and respiration of cells. Here, we have generated mice lacking exons 3 to 4 of the Pgc-1beta gene (Pgc-1beta(E3,4-/E3,4-) mice). These mice express a mutant protein that has reduced coactivation activity on a subset of transcription factors, including ERRalpha, a major target of PGC-1beta in the induction of mitochondrial gene expression. The mutant mice have reduced expression of OXPHOS genes and mitochondrial dysfunction in liver and skeletal muscle as well as elevated liver triglycerides. Euglycemic-hyperinsulinemic clamp and insulin signaling studies show that PGC-1beta mutant mice have normal skeletal muscle response to insulin but have hepatic insulin resistance. These results demonstrate that PGC-1beta is required for normal expression of OXPHOS genes and mitochondrial function in liver and skeletal muscle. Importantly, these abnormalities do not cause insulin resistance in skeletal muscle but cause substantially reduced insulin action in the liver.


Asunto(s)
Resistencia a la Insulina , Mitocondrias Hepáticas/metabolismo , Mitocondrias Musculares/metabolismo , Proteínas Mitocondriales/biosíntesis , Mutación , Transactivadores/metabolismo , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Técnica de Clampeo de la Glucosa , Hipoglucemiantes/farmacología , Insulina/farmacología , Resistencia a la Insulina/genética , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/patología , Mitocondrias Musculares/genética , Mitocondrias Musculares/patología , Proteínas Mitocondriales/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Especificidad de Órganos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Transactivadores/deficiencia , Factores de Transcripción , Receptor Relacionado con Estrógeno ERRalfa
16.
J Clin Invest ; 116(7): 1886-901, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16794735

RESUMEN

Leptin and insulin have been identified as fuel sensors acting in part through their hypothalamic receptors to inhibit food intake and stimulate energy expenditure. As their intracellular signaling converges at the PI3K pathway, we directly addressed the role of phosphatidylinositol3,4,5-trisphosphate-mediated (PIP3-mediated) signals in hypothalamic proopiomelanocortin (POMC) neurons by inactivating the gene for the PIP3 phosphatase Pten specifically in this cell type. Here we show that POMC-specific disruption of Pten resulted in hyperphagia and sexually dimorphic diet-sensitive obesity. Although leptin potently stimulated Stat3 phosphorylation in POMC neurons of POMC cell-restricted Pten knockout (PPKO) mice, it failed to significantly inhibit food intake in vivo. POMC neurons of PPKO mice showed a marked hyperpolarization and a reduction in basal firing rate due to increased ATP-sensitive potassium (KATP) channel activity. Leptin was not able to elicit electrical activity in PPKO POMC neurons, but application of the PI3K inhibitor LY294002 and the KATP blocker tolbutamide restored electrical activity and leptin-evoked firing of POMC neurons in these mice. Moreover, icv administration of tolbutamide abolished hyperphagia in PPKO mice. These data indicate that PIP3-mediated signals are critical regulators of the melanocortin system via modulation of KATP channels.


Asunto(s)
Neuronas/metabolismo , Obesidad , Fosfohidrolasa PTEN/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Canales de Potasio/metabolismo , Proopiomelanocortina/metabolismo , Sistemas de Mensajero Secundario/fisiología , Animales , Cromonas/metabolismo , Dieta , Ingestión de Alimentos/efectos de los fármacos , Femenino , Hipoglucemiantes/farmacología , Hipotálamo/citología , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Noqueados , Morfolinas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Tolbutamida/farmacología
17.
J Clin Invest ; 115(12): 3564-72, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16322794

RESUMEN

Ghrelin is the endogenous ligand for the growth hormone secretagogue receptor (GHSR; ghrelin receptor). Since its discovery, accumulating evidence has suggested that ghrelin may play a role in signaling and reversing states of energy insufficiency. For example, ghrelin levels rise following food deprivation, and ghrelin administration stimulates feeding and increases body weight and adiposity. However, recent loss-of-function studies have raised questions regarding the physiological significance of ghrelin in regulating these processes. Here, we present results of a study using a novel GHSR-null mouse model, in which ghrelin administration fails to acutely stimulate food intake or activate arcuate nucleus neurons. We show that when fed a high-fat diet, both female and male GHSR-null mice eat less food, store less of their consumed calories, preferentially utilize fat as an energy substrate, and accumulate less body weight and adiposity than control mice. Similar effects on body weight and adiposity were also observed in female, but not male, GHSR-null mice fed standard chow. GHSR deletion also affected locomotor activity and levels of glycemia. These findings support the hypothesis that ghrelin-responsive pathways are an important component of coordinated body weight control. Moreover, our data suggest that ghrelin signaling is required for development of the full phenotype of diet-induced obesity.


Asunto(s)
Dieta , Obesidad/genética , Hormonas Peptídicas/fisiología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Tejido Adiposo/metabolismo , Alelos , Análisis de Varianza , Alimentación Animal , Animales , Glucemia/metabolismo , Southern Blotting , Western Blotting , Composición Corporal , Peso Corporal , Cruzamientos Genéticos , ADN/metabolismo , Femenino , Eliminación de Gen , Predisposición Genética a la Enfermedad , Genotipo , Ghrelina , Heterocigoto , Homeostasis , Hiperglucemia/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Leptina/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Neuronas/metabolismo , Obesidad/metabolismo , Hormonas Peptídicas/química , Fenotipo , ARN Mensajero/metabolismo , Receptores de Ghrelina , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinación Genética , Transducción de Señal , Tinción con Nitrato de Plata , Factores de Tiempo
18.
Cell Metab ; 1(1): 63-72, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16054045

RESUMEN

Leptin is required for normal energy and glucose homeostasis. The hypothalamic arcuate nucleus (ARH) has been proposed as an important site of leptin action. To assess the physiological significance of leptin signaling in the ARH, we used mice homozygous for a FLPe-reactivatable, leptin receptor null allele (Lepr(neo/neo) mice). Similar to Lepr(db/db) mice, these mice are obese, hyperglycemic, hyperinsulinemic, infertile, and hypoactive. To selectively restore leptin signaling in the ARH, we generated an adeno-associated virus expressing FLPe-recombinase, which was delivered unilaterally into the hypothalamus using stereotaxic injections. We found that unilateral restoration of leptin signaling in the ARH of Lepr(neo/neo) mice leads to a modest decrease in body weight and food intake. In contrast, unilateral reactivation markedly improved hyperinsulinemia and normalized blood glucose levels and locomotor activity. These data demonstrate that leptin signaling in the ARH is sufficient for mediating leptin's effects on glucose homeostasis and locomotor activity.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Alelos , Animales , Composición Corporal , Peso Corporal , Núcleo Celular/metabolismo , ADN Nucleotidiltransferasas/metabolismo , Fertilidad , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis , Homocigoto , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Genéticos , Actividad Motora , Neuronas/metabolismo , Consumo de Oxígeno , Receptores de Superficie Celular/metabolismo , Receptores de Leptina , Transducción de Señal , Factores de Tiempo
19.
Cancer Res ; 65(11): 4728-38, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15930291

RESUMEN

Tumor cell invasion is a process regulated by integrins, matrix-degrading enzymes, and interactions with host tissue stromal cells. The ADAM family of proteins plays an important role in modulating various cellular responses. Here, we show that an alternatively spliced variant of ADAM9 is secreted by hepatic stellate cells and promotes carcinoma invasion. ADAM9-S induced a highly invasive phenotype in several human tumor cell lines in Matrigel assays, and the protease activity of ADAM9-S was required for invasion. ADAM9-S binds directly to alpha6beta4 and alpha2beta1 integrins on the surface of colon carcinoma cells through the disintegrin domain. ADAM9-S was also able to cleave laminin and promote invasion. Analysis of human liver metastases revealed that ADAM9 is expressed by stromal liver myofibroblasts, particularly those that are localized within the tumor stroma at the invasive front. These results emphasize the importance of tumor-stromal interactions in invasion and suggest that ADAM9-S can be an important determinant in the ability of cancer cells to invade and colonize the liver.


Asunto(s)
Comunicación Celular/fisiología , Neoplasias del Colon/patología , Desintegrinas/fisiología , Neoplasias Hepáticas Experimentales/secundario , Hígado/patología , Proteínas de la Membrana/fisiología , Metaloendopeptidasas/fisiología , Proteínas ADAM , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Colágeno , Desintegrinas/biosíntesis , Desintegrinas/metabolismo , Combinación de Medicamentos , Humanos , Integrina alfa2beta1/metabolismo , Integrina alfa6beta4/metabolismo , Laminina , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/biosíntesis , Metaloendopeptidasas/metabolismo , Ratones , Datos de Secuencia Molecular , Invasividad Neoplásica , Proteoglicanos , Células del Estroma/metabolismo , Células del Estroma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA