Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell ; 135(5): 813-24, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-19041747

RESUMEN

N-acylphosphatidylethanolamines (NAPEs) are a relatively abundant group of plasma lipids of unknown physiological significance. Here, we show that NAPEs are secreted into circulation from the small intestine in response to ingested fat and that systemic administration of the most abundant circulating NAPE, at physiologic doses, decreases food intake in rats without causing conditioned taste aversion. Furthermore, (14)C-radiolabeled NAPE enters the brain and is particularly concentrated in the hypothalamus, and intracerebroventricular infusions of nanomolar amounts of NAPE reduce food intake, collectively suggesting that its effects may be mediated through direct interactions with the central nervous system. Finally, chronic NAPE infusion results in a reduction of both food intake and body weight, suggesting that NAPE and long-acting NAPE analogs may be novel therapeutic targets for the treatment of obesity.


Asunto(s)
Regulación del Apetito , Fosfatidiletanolaminas/fisiología , Amidas , Animales , Peso Corporal , Grasas de la Dieta/metabolismo , Endocannabinoides , Etanolaminas , Hipotálamo/metabolismo , Intestino Delgado/metabolismo , Ratones , Ratones Obesos , Actividad Motora , Obesidad/metabolismo , Ácidos Palmíticos/metabolismo , Fosfatidiletanolaminas/sangre , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Espectrometría de Masas en Tándem
2.
Analyst ; 142(23): 4431-4437, 2017 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-29072717

RESUMEN

Nicotinamide adenine dinucleotide (NAD) and nicotinamide adenine dinucleotide phosphate (NADP) are conserved metabolic cofactors that mediate reduction-oxidation (redox) reactions throughout all domains of life. The diversity of synthetic routes and cellular processes involving the transfer of reducing equivalents to and from these cofactors makes the accurate quantitation and metabolic tracing of NAD(H) and NADP(H) of broad interest. However, current analytical techniques typically rely on standard curves that do not incorporate confounding effects of the sample matrix. We utilized the essential requirement of niacin and tryptophan for NAD synthesis in mammalian cells to devise a stable isotope labeling by essential nutrients in cell culture (SILEC) method for efficient labeling of intracellular NAD(H) and NADP(H) pools. Coupling this approach with detection by liquid chromatography-high resolution mass spectrometry (LC-HRMS), we demonstrate the utility of incorporating a [13C315N1]-nicotinamide moiety into a library of NAD-derived metabolites for use as internal standards in matrixed samples. Using a two-label system incorporating [13C315N1]-nicotinamide and [13C11]-tryptophan, we quantify the relative contribution of salvage and de novo NAD synthesis, respectively, in S. cerevisiae and HepG2 human hepatocellular carcinoma cells under basal conditions. As a further proof-of-principle, we demonstrate an improvement in the linear range for quantification of NAD and apply this method to analysis of NAD(H) in mouse liver. This method demonstrates the generalizability of SILEC, and provides a simple method for generating a library of stable isotope labeled internal standards for quantifying and tracing the metabolism of cellular and tissue NAD(H) and NADP(H).


Asunto(s)
Marcaje Isotópico , NADP/metabolismo , NAD/metabolismo , Animales , Células Cultivadas , Cromatografía Liquida , Células Hep G2 , Humanos , Hígado/metabolismo , Espectrometría de Masas , Ratones , Saccharomyces cerevisiae
3.
Adv Exp Med Biol ; 977: 51-57, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28685427

RESUMEN

Aging is the greatest risk factor for many diseases. Intracellular concentrations of nicotinamide adenine dinucleotide (NAD+) and the NAD+-coupled redox state have been proposed to moderate many aging-related processes, yet the specific mechanisms remain unclear. The concentration of NAD+ falls with age in skeletal muscle, yet there is no consensus on whether aging will increase or decrease the redox potential of NAD+/NADH. Oxidized flavin groups (Fp) (e.g. FAD, i.e., flavin adenine dinucleotide, contained in flavoproteins) and NADH are intrinsic fluorescent indicators of oxidation and reduction status of tissue, respectively. The redox ratio, i.e., the ratio of Fp to NADH, may be a surrogate indicator of the NAD+/NADH redox potential. In this study we used the Chance redox scanner (NADH/Fp fluorescence imaging at low temperature) to investigate the effect of aging on the redox state of mitochondria in skeletal muscles. The results showed that there are borderline significant differences in nominal concentrations of Fp and NADH, but not in the redox ratio s when comparing 3.5-month and 13-month old muscles of mice (n = 6). It may be necessary to increase the number of muscle samples and study mice of more advanced age.


Asunto(s)
Envejecimiento/metabolismo , Músculos/metabolismo , NAD/metabolismo , Imagen Óptica/métodos , Animales , Flavoproteínas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Oxidación-Reducción
4.
J Biol Chem ; 290(3): 1546-58, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25411251

RESUMEN

The NAD biosynthetic precursors nicotinamide mononucleotide and nicotinamide riboside are reported to confer resistance to metabolic defects induced by high fat feeding in part by promoting oxidative metabolism in skeletal muscle. Similar effects are obtained by germ line deletion of major NAD-consuming enzymes, suggesting that the bioavailability of NAD is limiting for maximal oxidative capacity. However, because of their systemic nature, the degree to which these interventions exert cell- or tissue-autonomous effects is unclear. Here, we report a tissue-specific approach to increase NAD biosynthesis only in muscle by overexpressing nicotinamide phosphoribosyltransferase, the rate-limiting enzyme in the salvage pathway that converts nicotinamide to NAD (mNAMPT mice). These mice display a ∼50% increase in skeletal muscle NAD levels, comparable with the effects of dietary NAD precursors, exercise regimens, or loss of poly(ADP-ribose) polymerases yet surprisingly do not exhibit changes in muscle mitochondrial biogenesis or mitochondrial function and are equally susceptible to the metabolic consequences of high fat feeding. We further report that chronic elevation of muscle NAD in vivo does not perturb the NAD/NADH redox ratio. These studies reveal for the first time the metabolic effects of tissue-specific increases in NAD synthesis and suggest that critical sites of action for supplemental NAD precursors reside outside of the heart and skeletal muscle.


Asunto(s)
Citocinas/metabolismo , Músculo Esquelético/metabolismo , NAD/biosíntesis , Nicotinamida Fosforribosiltransferasa/metabolismo , Oxígeno/metabolismo , Animales , Sitios de Unión , Calorimetría , Cromatografía Líquida de Alta Presión , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Musculares/metabolismo , NAD/metabolismo , Mononucleótido de Nicotinamida/metabolismo , Oxidación-Reducción , Poli(ADP-Ribosa) Polimerasas/metabolismo
5.
J Biol Chem ; 286(42): 36163-70, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21832042

RESUMEN

Recent studies have implicated endoplasmic reticulum (ER) stress in insulin resistance associated with caloric excess. In mice placed on a 3-day high fat diet, we find augmented eIF2α signaling, together with hepatic lipid accumulation and insulin resistance. To clarify the role of the liver ER stress-dependent phospho-eIF2α (eIF2α-P) pathway in response to acute caloric excess on liver and muscle glucose and lipid metabolism, we studied transgenic mice in which the hepatic ER stress-dependent eIF2α-P pathway was inhibited by overexpressing a constitutively active C-terminal fragment of GADD34/PPP1R15a, a regulatory subunit of phosphatase that terminates ER stress signaling by phospho-eIF2α. Inhibition of the eIF2α-P signaling in liver led to a decrease in hepatic glucose production in the basal and clamped state, which could be attributed to reduced gluconeogenic gene expression, resulting in reduced basal plasma glucose concentrations. Surprisingly, hepatic eIF2α inhibition also impaired insulin-stimulated muscle and adipose tissue insulin sensitivity. This latter effect could be attributed at least in part by an increase in circulating IGFBP-3 levels in the transgenic animals. In addition, infusion of insulin during a hyperinsulinemic-euglycemic clamp induced conspicuous ER stress in the 3-day high fat diet-fed mice, which was aggravated through continuous dephosphorylation of eIF2α. Together, these data imply that the hepatic ER stress eIF2α signaling pathway affects hepatic glucose production without altering hepatic insulin sensitivity. Moreover, hepatic ER stress-dependent eIF2α-P signaling is implicated in an unanticipated cross-talk between the liver and peripheral organs to influence insulin sensitivity, probably via IGFBP-3. Finally, eIF2α is crucial for proper resolution of insulin-induced ER stress.


Asunto(s)
Tejido Adiposo/metabolismo , Retículo Endoplásmico/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Músculo Esquelético/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/genética , Insulina/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Transgénicos , Fosforilación/fisiología , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Transducción de Señal/fisiología
6.
Hepatology ; 54(5): 1650-60, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21793029

RESUMEN

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) and insulin resistance have recently been found to be associated with increased plasma concentrations of apolipoprotein CIII (APOC3) in humans carrying single nucleotide polymorphisms within the insulin response element of the APOC3 gene. To examine whether increased expression of APOC3 would predispose mice to NAFLD and hepatic insulin resistance, human APOC3 overexpressing (ApoC3Tg) mice were metabolically phenotyped following either a regular chow or high-fat diet (HFD). After HFD feeding, ApoC3Tg mice had increased hepatic triglyceride accumulation, which was associated with cellular ballooning and inflammatory changes. ApoC3Tg mice also manifested severe hepatic insulin resistance assessed by a hyperinsulinemic-euglycemic clamp, which could mostly be attributed to increased hepatic diacylglycerol content, protein kinase C-ϵ activation, and decreased insulin-stimulated Akt2 activity. Increased hepatic triglyceride content in the HFD-fed ApoC3Tg mice could be attributed to a ≈ 70% increase in hepatic triglyceride uptake and ≈ 50% reduction hepatic triglyceride secretion. CONCLUSION: These data demonstrate that increase plasma APOC3 concentrations predispose mice to diet-induced NAFLD and hepatic insulin resistance.


Asunto(s)
Apolipoproteína C-III/sangre , Apolipoproteína C-III/genética , Hígado Graso/genética , Hígado Graso/metabolismo , Resistencia a la Insulina/genética , Alimentación Animal , Animales , Apolipoproteína B-100/metabolismo , Glucemia/metabolismo , VLDL-Colesterol/metabolismo , Grasas de la Dieta/farmacología , Diglicéridos/metabolismo , Femenino , Predisposición Genética a la Enfermedad/genética , Hiperinsulinismo/metabolismo , Masculino , Ratones , Ratones Mutantes , Enfermedad del Hígado Graso no Alcohólico , Periodo Posprandial/fisiología , Proteína Quinasa C/metabolismo , Triglicéridos/metabolismo , Triglicéridos/farmacocinética
7.
Am J Physiol Endocrinol Metab ; 299(5): E808-15, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20807839

RESUMEN

Low-carbohydrate, high-fat ketogenic diets (KD) have been suggested to be more effective in promoting weight loss than conventional caloric restriction, whereas their effect on hepatic glucose and lipid metabolism and the mechanisms by which they may promote weight loss remain controversial. The aim of this study was to explore the role of KD on liver and muscle insulin sensitivity, hepatic lipid metabolism, energy expenditure, and food intake. Using hyperinsulinemic-euglycemic clamps, we studied insulin action in mice fed a KD or regular chow (RC). Body composition was assessed by ¹H magnetic resonance spectroscopy. Despite being 15% lighter (P < 0.001) than RC-fed mice because of a 17% increase in energy expenditure (P < 0.001), KD-fed mice manifested severe hepatic insulin resistance, as reflected by decreased suppression (0% vs. 100% in RC-fed mice, P < 0.01) of endogenous glucose production during the clamp. Hepatic insulin resistance could be attributed to a 350% increase in hepatic diacylglycerol content (P < 0.001), resulting in increased activation of PKCε (P < 0.05) and decreased insulin receptor substrate-2 tyrosine phosphorylation (P < 0.01). Food intake was 56% (P < 0.001) lower in KD-fed mice, despite similar caloric intake, and could partly be attributed to a more than threefold increase (P < 0.05) in plasma N-acylphosphatidylethanolamine concentrations. In conclusion, despite preventing weight gain in mice, KD induces hepatic insulin resistance secondary to increased hepatic diacylglycerol content. Given the key role of nonalcoholic fatty liver disease in the development of type 2 diabetes and the widespread use of KD for the treatment of obesity, these results may have potentially important clinical implications.


Asunto(s)
Grasas de la Dieta/metabolismo , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Músculo Esquelético/metabolismo , Animales , Composición Corporal/fisiología , Técnica de Clampeo de la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Skelet Muscle ; 10(1): 30, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-33092650

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disorder stemming from a loss of functional dystrophin. Current therapeutic options for DMD are limited, as small molecule modalities remain largely unable to decrease the incidence or mitigate the consequences of repetitive mechanical insults to the muscle during eccentric contractions (ECCs). METHODS: Using a metabolomics-based approach, we observed distinct and transient molecular phenotypes in muscles of dystrophin-deficient MDX mice subjected to ECCs. Among the most chronically depleted metabolites was nicotinamide adenine dinucleotide (NAD), an essential metabolic cofactor suggested to protect muscle from structural and metabolic degeneration over time. We tested whether the MDX muscle NAD pool can be expanded for therapeutic benefit using two complementary small molecule strategies: provision of a biosynthetic precursor, nicotinamide riboside, or specific inhibition of the NAD-degrading ADP-ribosyl cyclase, CD38. RESULTS: Administering a novel, potent, and orally available CD38 antagonist to MDX mice successfully reverted a majority of the muscle metabolome toward the wildtype state, with a pronounced impact on intermediates of the pentose phosphate pathway, while supplementing nicotinamide riboside did not significantly affect the molecular phenotype of the muscle. However, neither strategy sustainably increased the bulk tissue NAD pool, lessened muscle damage markers, nor improved maximal hindlimb strength following repeated rounds of eccentric challenge and recovery. CONCLUSIONS: In the absence of dystrophin, eccentric injury contributes to chronic intramuscular NAD depletion with broad pleiotropic effects on the molecular phenotype of the tissue. These molecular consequences can be more effectively overcome by inhibiting the enzymatic activity of CD38 than by supplementing nicotinamide riboside. However, we found no evidence that either small molecule strategy is sufficient to restore muscle contractile function or confer protection from eccentric injury, undermining the modulation of NAD metabolism as a therapeutic approach for DMD.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Metaboloma , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/tratamiento farmacológico , NAD/metabolismo , Niacinamida/análogos & derivados , Compuestos de Piridinio/farmacología , ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , Animales , Distrofina/deficiencia , Inhibidores Enzimáticos/uso terapéutico , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Contracción Muscular , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Niacinamida/farmacología , Niacinamida/uso terapéutico , Compuestos de Piridinio/uso terapéutico
9.
Aging (Albany NY) ; 12(18): 17786-17799, 2020 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-32960787

RESUMEN

Rapamycin delays multiple age-related conditions and extends lifespan in organisms ranging from yeast to mice. However, the mechanisms by which rapamycin influences longevity are incompletely understood. The objective of this study was to investigate the effect of rapamycin on NAD+/NADH redox balance. We report that the NAD+/NADH ratio of C2C12 myoblasts or differentiated myotubes significantly decreases over time in culture, and that rapamycin prevents this effect. Despite lowering the NADH available to support ATP generation, rapamycin increases ATP availability, consistent with lowering energetic demand. Although rapamycin did not change the NAD+/NADH ratio or steady-state ATP concentration in the livers, kidneys, or muscles of young mice, optical redox imaging revealed that rapamycin caused a substantial decline in the NADH content and an increase in the optical redox ratio (a surrogate of NAD+/NADH redox ratio) in muscles from aged mice. Collectively, these data suggest that rapamycin favors a more oxidized NAD+/NADH ratio in aged muscle, which may influence metabolism and the activity of NAD+-dependent enzymes. This study provides new insight into the mechanisms by which rapamycin might influence the aging process to improve health and longevity among the aging population.

10.
Cell Metab ; 27(5): 1067-1080.e5, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29685734

RESUMEN

The redox cofactor nicotinamide adenine dinucleotide (NAD) plays a central role in metabolism and is a substrate for signaling enzymes including poly-ADP-ribose-polymerases (PARPs) and sirtuins. NAD concentration falls during aging, which has triggered intense interest in strategies to boost NAD levels. A limitation in understanding NAD metabolism has been reliance on concentration measurements. Here, we present isotope-tracer methods for NAD flux quantitation. In cell lines, NAD was made from nicotinamide and consumed largely by PARPs and sirtuins. In vivo, NAD was made from tryptophan selectively in the liver, which then excreted nicotinamide. NAD fluxes varied widely across tissues, with high flux in the small intestine and spleen and low flux in the skeletal muscle. Intravenous administration of nicotinamide riboside or mononucleotide delivered intact molecules to multiple tissues, but the same agents given orally were metabolized to nicotinamide in the liver. Thus, flux analysis can reveal tissue-specific NAD metabolism.


Asunto(s)
Hígado/metabolismo , NAD/análisis , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Sirtuinas/metabolismo , Triptófano/metabolismo , Animales , Femenino , Células HCT116 , Células Hep G2 , Humanos , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , NAD/biosíntesis , Niacinamida/administración & dosificación , Niacinamida/farmacocinética , Bazo/metabolismo
11.
Cell Metab ; 27(3): 667-676.e4, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29514072

RESUMEN

The role in longevity and healthspan of nicotinamide (NAM), the physiological precursor of NAD+, is elusive. Here, we report that chronic NAM supplementation improves healthspan measures in mice without extending lifespan. Untargeted metabolite profiling of the liver and metabolic flux analysis of liver-derived cells revealed NAM-mediated improvement in glucose homeostasis in mice on a high-fat diet (HFD) that was associated with reduced hepatic steatosis and inflammation concomitant with increased glycogen deposition and flux through the pentose phosphate and glycolytic pathways. Targeted NAD metabolome analysis in liver revealed depressed expression of NAM salvage in NAM-treated mice, an effect counteracted by higher expression of de novo NAD biosynthetic enzymes. Although neither hepatic NAD+ nor NADP+ was boosted by NAM, acetylation of some SIRT1 targets was enhanced by NAM supplementation in a diet- and NAM dose-dependent manner. Collectively, our results show health improvement in NAM-supplemented HFD-fed mice in the absence of survival effects.


Asunto(s)
Suplementos Dietéticos , Envejecimiento Saludable/metabolismo , Hígado , NAD/metabolismo , Niacinamida/farmacología , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hígado Graso/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Hígado/efectos de los fármacos , Hígado/metabolismo , Longevidad , Ratones Endogámicos C57BL , Niacinamida/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Sirtuina 1/metabolismo
12.
Diabetes ; 65(4): 927-41, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26858361

RESUMEN

Rapamycin extends life span in mice, yet paradoxically causes lipid dysregulation and glucose intolerance through mechanisms that remain incompletely understood. Whole-body energy balance can be influenced by beige/brite adipocytes, which are inducible by cold and other stimuli via ß-adrenergic signaling in white adipose depots. Induction of beige adipocytes is considered a promising strategy to combat obesity because of their ability to metabolize glucose and lipids, dissipating the resulting energy as heat through uncoupling protein 1. Here, we report that rapamycin blocks the ability of ß-adrenergic signaling to induce beige adipocytes and expression of thermogenic genes in white adipose depots. Rapamycin enhanced transcriptional negative feedback on the ß3-adrenergic receptor. However, thermogenic gene expression remained impaired even when the receptor was bypassed with a cell-permeable cAMP analog, revealing the existence of a second inhibitory mechanism. Accordingly, rapamycin-treated mice are cold intolerant, failing to maintain body temperature and weight when shifted to 4°C. Adipocyte-specific deletion of the mTORC1 subunit Raptor recapitulated the block in ß-adrenergic signaling. Our findings demonstrate a positive role for mTORC1 in the recruitment of beige adipocytes and suggest that inhibition of ß-adrenergic signaling by rapamycin may contribute to its physiological effects.


Asunto(s)
Tejido Adiposo Blanco/efectos de los fármacos , Sirolimus/farmacología , Termogénesis/efectos de los fármacos , Adipocitos/efectos de los fármacos , Adipocitos/fisiología , Tejido Adiposo Blanco/metabolismo , Animales , Dioxoles/farmacología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Termogénesis/genética
13.
Cell Metab ; 24(2): 269-82, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27508874

RESUMEN

NAD is an obligate co-factor for the catabolism of metabolic fuels in all cell types. However, the availability of NAD in several tissues can become limited during genotoxic stress and the course of natural aging. The point at which NAD restriction imposes functional limitations on tissue physiology remains unknown. We examined this question in murine skeletal muscle by specifically depleting Nampt, an essential enzyme in the NAD salvage pathway. Knockout mice exhibited a dramatic 85% decline in intramuscular NAD content, accompanied by fiber degeneration and progressive loss of both muscle strength and treadmill endurance. Administration of the NAD precursor nicotinamide riboside rapidly ameliorated functional deficits and restored muscle mass despite having only a modest effect on the intramuscular NAD pool. Additionally, lifelong overexpression of Nampt preserved muscle NAD levels and exercise capacity in aged mice, supporting a critical role for tissue-autonomous NAD homeostasis in maintaining muscle mass and function.


Asunto(s)
Homeostasis , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , NAD/metabolismo , Administración Oral , Envejecimiento/fisiología , Animales , Disponibilidad Biológica , Metabolismo Energético , Glucosa/metabolismo , Inflamación/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Fuerza Muscular , Músculo Esquelético/enzimología , Músculo Esquelético/fisiopatología , Necrosis , Niacinamida/administración & dosificación , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Niacinamida/farmacología , Nicotinamida Fosforribosiltransferasa/deficiencia , Nicotinamida Fosforribosiltransferasa/metabolismo , Tamaño de los Órganos , Condicionamiento Físico Animal , Compuestos de Piridinio , Transcripción Genética
14.
Cell Metab ; 23(6): 1093-1112, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27304509

RESUMEN

Calorie restriction (CR) is the most robust non-genetic intervention to delay aging. However, there are a number of emerging experimental variables that alter CR responses. We investigated the role of sex, strain, and level of CR on health and survival in mice. CR did not always correlate with lifespan extension, although it consistently improved health across strains and sexes. Transcriptional and metabolomics changes driven by CR in liver indicated anaplerotic filling of the Krebs cycle together with fatty acid fueling of mitochondria. CR prevented age-associated decline in the liver proteostasis network while increasing mitochondrial number, preserving mitochondrial ultrastructure and function with age. Abrogation of mitochondrial function negated life-prolonging effects of CR in yeast and worms. Our data illustrate the complexity of CR in the context of aging, with a clear separation of outcomes related to health and survival, highlighting complexities of translation of CR into human interventions.


Asunto(s)
Envejecimiento/metabolismo , Ingestión de Energía , Caracteres Sexuales , Envejecimiento/genética , Animales , Autofagia/genética , Biomarcadores/metabolismo , Restricción Calórica , Análisis por Conglomerados , Ingestión de Energía/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Homeostasis/genética , Sulfuro de Hidrógeno/metabolismo , Islotes Pancreáticos/anatomía & histología , Hígado/metabolismo , Hígado/ultraestructura , Longevidad/genética , Longevidad/fisiología , Masculino , Metaboloma , Metabolómica , Ratones , Ratones Endogámicos , Mitocondrias/metabolismo , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo
15.
Mol Metab ; 4(12): 926-39, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26909309

RESUMEN

OBJECTIVES: Hyperglycemia and elevated blood lipids are the presumed precipitating causes of ß-cell damage in T2DM as the result of a process termed "glucolipotoxicity". Here, we tested whether glucolipotoxic pathophysiology is caused by defective bioenergetics using islets in culture. METHODS: Insulin secretion, respiration, ATP generation, fatty acid (FA) metabolite profiles and gene expression were determined in isolated islets treated under glucolipotoxic culture conditions. RESULTS: Over time, chronic exposure of mouse islets to FAs with glucose leads to bioenergetic failure and reduced insulin secretion upon stimulation with glucose or amino acids. Islets exposed to glucolipotoxic conditions displayed biphasic changes of the oxygen consumption rate (OCR): an initial increase in baseline and Vmax of OCR after 3 days, followed by decreased baseline and glucose stimulated OCR after 5 days. These changes were associated with lower islet ATP levels, impaired glucose-induced ATP generation, a trend for reduced mitochondrial DNA content and reduced expression of mitochondrial transcription factor A (Tfam). We discovered the accumulation of carnitine esters of hydroxylated long chain FAs, in particular 3-hydroxytetradecenoyl-carnitine. CONCLUSIONS: As long chain 3-hydroxylated FA metabolites are known to uncouple heart and brain mitochondria [53], [54], [55], we propose that under glucolipotoxic condition, unsaturated hydroxylated long-chain FAs accumulate, uncouple and ultimately inhibit ß-cell respiration. This leads to the slow deterioration of mitochondrial function progressing to bioenergetics ß-cell failure.

16.
Diabetes ; 60(3): 890-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21357472

RESUMEN

OBJECTIVE: Inhibition of the Na(+)-glucose cotransporter type 2 (SGLT2) is currently being pursued as an insulin-independent treatment for diabetes; however, the behavioral and metabolic consequences of SGLT2 deletion are unknown. Here, we used a SGLT2 knockout mouse to investigate the effect of increased renal glucose excretion on glucose homeostasis, insulin sensitivity, and pancreatic ß-cell function. RESEARCH DESIGN AND METHODS: SGLT2 knockout mice were fed regular chow or a high-fat diet (HFD) for 4 weeks, or backcrossed onto the db/db background. The analysis used metabolic cages, glucose tolerance tests, euglycemic and hyperglycemic clamps, as well as isolated islet and perifusion studies. RESULTS: SGLT2 deletion resulted in a threefold increase in urine output and a 500-fold increase in glucosuria, as well as compensatory increases in feeding, drinking, and activity. SGLT2 knockout mice were protected from HFD-induced hyperglycemia and glucose intolerance and had reduced plasma insulin concentrations compared with controls. On the db/db background, SGLT2 deletion prevented fasting hyperglycemia, and plasma insulin levels were also dramatically improved. Strikingly, prevention of hyperglycemia by SGLT2 knockout in db/db mice preserved pancreatic ß-cell function in vivo, which was associated with a 60% increase in ß-cell mass and reduced incidence of ß-cell death. CONCLUSIONS: Prevention of renal glucose reabsorption by SGLT2 deletion reduced HFD- and obesity-associated hyperglycemia, improved glucose intolerance, and increased glucose-stimulated insulin secretion in vivo. Taken together, these data support SGLT2 inhibition as a viable insulin-independent treatment of type 2 diabetes.


Asunto(s)
Glucosa/metabolismo , Homeostasis/genética , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Análisis de Varianza , Animales , Apoptosis/genética , Grasas de la Dieta/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Insulina/sangre , Resistencia a la Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/fisiopatología , Riñón/metabolismo , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/fisiopatología , Transportador 2 de Sodio-Glucosa/genética
17.
Diabetes ; 60(12): 3235-45, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22110092

RESUMEN

OBJECTIVE: Macrophage recruitment to adipose tissue is a reproducible feature of obesity. However, the events that result in chemokine production and macrophage recruitment to adipose tissue during states of energetic excess are not clear. Sirtuin 1 (SirT1) is an essential nutrient-sensing histone deacetylase, which is increased by caloric restriction and reduced by overfeeding. We discovered that SirT1 depletion causes anorexia by stimulating production of inflammatory factors in white adipose tissue and thus posit that decreases in SirT1 link overnutrition and adipose tissue inflammation. RESEARCH DESIGN AND METHODS: We used antisense oligonucleotides to reduce SirT1 to levels similar to those seen during overnutrition and studied SirT1-overexpressing transgenic mice and fat-specific SirT1 knockout animals. Finally, we analyzed subcutaneous adipose tissue biopsies from two independent cohorts of human subjects. RESULTS: We found that inducible or genetic reduction of SirT1 in vivo causes macrophage recruitment to adipose tissue, whereas overexpression of SirT1 prevents adipose tissue macrophage accumulation caused by chronic high-fat feeding. We also found that SirT1 expression in human subcutaneous fat is inversely related to adipose tissue macrophage infiltration. CONCLUSIONS: Reduction of adipose tissue SirT1 expression, which leads to histone hyperacetylation and ectopic inflammatory gene expression, is identified as a key regulatory component of macrophage influx into adipose tissue during overnutrition in rodents and humans. Our results suggest that SirT1 regulates adipose tissue inflammation by controlling the gain of proinflammatory transcription in response to inducers such as fatty acids, hypoxia, and endoplasmic reticulum stress.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Sirtuina 1/inmunología , Sirtuina 1/metabolismo , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/metabolismo , Animales , Índice de Masa Corporal , Grasas de la Dieta/efectos adversos , Citometría de Flujo , Humanos , Técnicas In Vitro , Inflamación/inducido químicamente , Inflamación/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirtuina 1/genética , Factor de Necrosis Tumoral alfa/metabolismo , Pérdida de Peso/genética , Pérdida de Peso/fisiología
18.
Cell Metab ; 14(2): 184-95, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21803289

RESUMEN

Reduced expression of the Indy (I'm Not Dead, Yet) gene in D. melanogaster and its homolog in C. elegans prolongs life span and in D. melanogaster augments mitochondrial biogenesis in a manner akin to caloric restriction. However, the cellular mechanism by which Indy does this is unknown. Here, we report on the knockout mouse model of the mammalian Indy (mIndy) homolog, SLC13A5. Deletion of mIndy in mice (mINDY(-/-) mice) reduces hepatocellular ATP/ADP ratio, activates hepatic AMPK, induces PGC-1α, inhibits ACC-2, and reduces SREBP-1c levels. This signaling network promotes hepatic mitochondrial biogenesis, lipid oxidation, and energy expenditure and attenuates hepatic de novo lipogenesis. Together, these traits protect mINDY(-/-) mice from the adiposity and insulin resistance that evolve with high-fat feeding and aging. Our studies demonstrate a profound effect of mIndy on mammalian energy metabolism and suggest that mINDY might be a therapeutic target for the treatment of obesity and type 2 diabetes.


Asunto(s)
Adiposidad/genética , Metabolismo Energético/genética , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/genética , Simportadores/biosíntesis , Envejecimiento , Animales , Restricción Calórica , Transportadores de Ácidos Dicarboxílicos , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Obesidad/genética , Simportadores/deficiencia , Simportadores/genética
19.
Diabetes ; 59(6): 1424-34, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20299477

RESUMEN

OBJECTIVE: Thioredoxin interacting protein (Txnip), a regulator of cellular oxidative stress, is induced by hyperglycemia and inhibits glucose uptake into fat and muscle, suggesting a role for Txnip in type 2 diabetes pathogenesis. Here, we tested the hypothesis that Txnip-null (knockout) mice are protected from insulin resistance induced by a high-fat diet. RESEARCH DESIGN AND METHODS: Txnip gene-deleted (knockout) mice and age-matched wild-type littermate control mice were maintained on a standard chow diet or subjected to 4 weeks of high-fat feeding. Mice were assessed for body composition, fat development, energy balance, and insulin responsiveness. Adipogenesis was measured from ex vivo fat preparations, and in mouse embryonic fibroblasts (MEFs) and 3T3-L1 preadipocytes after forced manipulation of Txnip expression. RESULTS: Txnip knockout mice gained significantly more adipose mass than controls due to a primary increase in both calorie consumption and adipogenesis. Despite increased fat mass, Txnip knockout mice were markedly more insulin sensitive than controls, and augmented glucose transport was identified in both adipose and skeletal muscle. RNA interference gene-silenced preadipocytes and Txnip(-/-) MEFs were markedly adipogenic, whereas Txnip overexpression impaired adipocyte differentiation. As increased adipogenesis and insulin sensitivity suggested aspects of augmented peroxisome proliferator-activated receptor-gamma (PPARgamma) response, we investigated Txnip's regulation of PPARgamma function; manipulation of Txnip expression directly regulated PPARgamma expression and activity. CONCLUSIONS: Txnip deletion promotes adiposity in the face of high-fat caloric excess; however, loss of this alpha-arrestin protein simultaneously enhances insulin responsiveness in fat and skeletal muscle, revealing Txnip as a novel mediator of insulin resistance and a regulator of adipogenesis.


Asunto(s)
Adiposidad/genética , Proteínas Portadoras/genética , Eliminación de Gen , Insulina/fisiología , Tiorredoxinas/genética , Células 3T3/fisiología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Grasas de la Dieta , Regulación de la Expresión Génica , Técnica de Clampeo de la Glucosa , Humanos , Hiperinsulinismo , Insulina/farmacología , Resistencia a la Insulina/genética , Ratones , Ratones Noqueados , Obesidad/genética , Estrés Oxidativo/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA