Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Metabolism ; 152: 155765, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38142958

RESUMEN

BACKGROUND AND AIM: The excessive accumulation of lipid droplets (LDs) is a defining characteristic of nonalcoholic fatty liver disease (NAFLD). The interaction between LDs and mitochondria is functionally important for lipid metabolism homeostasis. Exercise improves NAFLD, but it is not known if it has an effect on hepatic LD-mitochondria interactions. Here, we investigated the influence of exercise on LD-mitochondria interactions and its significance in the context of NAFLD. APPROACH AND RESULTS: Mice were fed high-fat diet (HFD) or HFD-0.1 % methionine and choline-deficient diet (MCD) to emulate simple hepatic steatosis or non-alcoholic steatohepatitis, respectively. In both models, aerobic exercise decreased the size of LDs bound to mitochondria and the number of LD-mitochondria contacts. Analysis showed that the effects of exercise on HOMA-IR and liver triglyceride levels were independent of changes in body weight, and a positive correlation was observed between the number of LD-mitochondria contacts and NAFLD severity and with the lipid droplet size bound to mitochondria. Cellular fractionation studies revealed that ATP-coupled respiration and fatty acid oxidation (FAO) were greater in hepatic peridroplet mitochondria (PDM) from HFD-fed exercised mice than from equivalent sedentary mice. Finally, exercise increased FAO and mitofusin-2 abundance exclusively in PDM through a mechanism involving the curvature of mitochondrial membranes and the abundance of saturated lipids. Accordingly, hepatic mitofusin-2 ablation prevented exercise-induced FAO in PDM. CONCLUSIONS: This study demonstrates that aerobic exercise has beneficial effects in murine NAFLD models by lessening the interactions between hepatic LDs and mitochondria, and by decreasing LD size, correlating with a reduced severity of NAFLD. Additionally, aerobic exercise increases FAO in PDM and this process is reliant on Mfn-2 enrichment, which modifies LD-mitochondria communication.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Masculino , Ratones , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo
2.
Int J Mol Sci ; 24(3)2023 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-36768897

RESUMEN

Many lines of evidence demonstrate a correlation between liver glycogen content and food intake. We previously demonstrated that mice overexpressing protein targeting to glycogen (PTG) specifically in the liver-which have increased glycogen content in this organ-are protected from high-fat diet (HFD)-induced obesity by reduced food intake. However, the use of PTG to increase liver glycogen implies certain limitations. PTG stimulates glycogen synthesis but also inhibits the enzyme responsible for glycogen degradation. Furthermore, as PTG is a regulatory subunit of protein phosphatase 1 (PP1), which regulates many cellular functions, its overexpression could have side effects beyond the regulation of glycogen metabolism. Therefore, it is necessary to determine whether the direct activation of glycogen synthesis, without affecting its degradation or other cellular functions, has the same effects. To this end, we generated mice overexpressing a non-inactivatable form of glycogen synthase (GS) specifically in the liver (9A-MGSAlb mice). Control and 9a-MGSAlb mice were fed a standard diet (SD) or HFD for 16 weeks. Glucose tolerance and feeding behavior were analyzed. 9A-MGSAlb mice showed an increase in hepatic glycogen in fed and fasting conditions. When fed an HFD, these animals preserved their hepatic energy state, had a reduced food intake, and presented a lower body weight and fat mass than control animals, without changes in energy expenditure. Furthermore, 9A-MGSAlb animals showed improved glucose tolerance when fed an SD or HFD. Moreover, liver triacylglycerol levels that were increased after HFD feeding were lower in these mice. These results confirm that increased liver glycogen stores contribute to decreased appetite and improve glucose tolerance in mice fed an HFD. On the basis of our findings, strategies to preserve hepatic glycogen stores emerge as potential treatments for obesity and hyperglycemia.


Asunto(s)
Intolerancia a la Glucosa , Glucógeno Hepático , Animales , Ratones , Peso Corporal , Dieta Alta en Grasa , Ingestión de Alimentos/fisiología , Glucosa/metabolismo , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/prevención & control , Intolerancia a la Glucosa/metabolismo , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/prevención & control , Obesidad/metabolismo
3.
Redox Biol ; 61: 102630, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36796135

RESUMEN

Type 2 diabetes mellitus (T2D) affects millions of people worldwide and is one of the leading causes of morbidity and mortality. The skeletal muscle (SKM) is one of the most important tissues involved in maintaining glucose homeostasis and substrate oxidation, and it undergoes insulin resistance in T2D. In this study, we identify the existence of alterations in the expression of mitochondrial aminoacyl-tRNA synthetases (mt-aaRSs) in skeletal muscle from two different forms of T2D: early-onset type 2 diabetes (YT2) (onset of the disease before 30 years of age) and the classical form of the disease (OT2). GSEA analysis from microarray studies revealed the repression of mitochondrial mt-aaRSs independently of age, which was validated by real-time PCR assays. In agreement with this, a reduced expression of several encoding mt-aaRSs was also detected in skeletal muscle from diabetic (db/db) mice but not in obese ob/ob mice. In addition, the expression of the mt-aaRSs proteins most relevant in the synthesis of mitochondrial proteins, threonyl-tRNA, and leucyl-tRNA synthetases (TARS2 and LARS2) were also repressed in muscle from db/db mice. It is likely that these alterations participate in the reduced expression of proteins synthesized in the mitochondria detected in db/db mice. We also document an increased iNOS abundance in mitochondrial-enriched muscle fractions from diabetic mice that may inhibit aminoacylation of TARS2 and LARS2 by nitrosative stress. Our results indicate a reduced expression of mt-aaRSs in skeletal muscle from T2D patients, which may participate in the reduced expression of proteins synthesized in mitochondria. An enhanced mitochondrial iNOS could play a regulatory role in diabetes.


Asunto(s)
Aminoacil-ARNt Sintetasas , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Ratones , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo , Aminoacil-ARNt Sintetasas/genética , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , ARN de Transferencia/metabolismo
4.
Front Endocrinol (Lausanne) ; 13: 969924, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36157460

RESUMEN

Increased liver glycogen content has been shown to reduce food intake, attenuate obesity, and improve glucose tolerance in a mouse model of high-fat diet (HFD)-induced obesity. Here we studied the contribution of liver glycogen to the regulation of obesity and glucose metabolism in a model of type 2 diabetes and obesity, namely the db/db mouse. To this end, we crossed db/db mice with animals overexpressing protein targeting to glycogen (PTG) in the liver to generate db/db mice with increased liver glycogen content (db/db-PTG). Hepatic PTG overexpression reduced food intake and fat weight and attenuated obesity and hyperglycemia in db/db mice. Db/db-PTG mice showed similar energy expenditure and physical activity to db/db mice. PTG overexpression reduced liver phosphoenolpyruvate carboxykinase (PEPCK) protein levels and repressed hepatic glucose production in db/db mice. Moreover, increased liver glycogen elevated hepatic ATP content in these animals. However, lipid metabolism was not modified by PTG overexpression. In conclusion, increased liver glycogen content ameliorates the diabetic and obesity phenotype in db/db mice.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperglucemia , Adenosina Trifosfato/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Hiperglucemia/metabolismo , Hiperglucemia/prevención & control , Lípidos , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Ratones , Obesidad/metabolismo , Fosfoenolpiruvato/metabolismo
5.
Mol Neurobiol ; 59(2): 1214-1229, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34962634

RESUMEN

Lafora disease (LD) is a fatal childhood-onset dementia characterized by the extensive accumulation of glycogen aggregates-the so-called Lafora Bodies (LBs)-in several organs. The accumulation of LBs in the brain underlies the neurological phenotype of the disease. LBs are composed of abnormal glycogen and various associated proteins, including p62, an autophagy adaptor that participates in the aggregation and clearance of misfolded proteins. To study the role of p62 in the formation of LBs and its participation in the pathology of LD, we generated a mouse model of the disease (malinKO) lacking p62. Deletion of p62 prevented LB accumulation in skeletal muscle and cardiac tissue. In the brain, the absence of p62 altered LB morphology and increased susceptibility to epilepsy. These results demonstrate that p62 participates in the formation of LBs and suggest that the sequestration of abnormal glycogen into LBs is a protective mechanism through which it reduces the deleterious consequences of its accumulation in the brain.


Asunto(s)
Enfermedad de Lafora , Animales , Modelos Animales de Enfermedad , Glucógeno/metabolismo , Cuerpos de Inclusión/metabolismo , Enfermedad de Lafora/genética , Ratones , Ratones Noqueados , Proteína Sequestosoma-1
6.
J Biol Chem ; 297(2): 100976, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34284060

RESUMEN

Muscle glycogen depletion has been proposed as one of the main causes of fatigue during exercise. However, few studies have addressed the contribution of liver glycogen to exercise performance. Using a low-intensity running protocol, here, we analyzed exercise capacity in mice overexpressing protein targeting to glycogen (PTG) specifically in the liver (PTGOE mice), which show a high concentration of glycogen in this organ. PTGOE mice showed improved exercise capacity, as determined by the distance covered and time ran in an extenuating endurance exercise, compared with control mice. Moreover, fasting decreased exercise capacity in control mice but not in PTGOE mice. After exercise, liver glycogen stores were totally depleted in control mice, but PTGOE mice maintained significant glycogen levels even in fasting conditions. In addition, PTGOE mice displayed an increased hepatic energy state after exercise compared with control mice. Exercise caused a reduction in the blood glucose concentration in control mice that was less pronounced in PTGOE mice. No changes were found in the levels of blood lactate, plasma free fatty acids, or ß-hydroxybutyrate. Plasma glucagon was elevated after exercise in control mice, but not in PTGOE mice. Exercise-induced changes in skeletal muscle were similar in both genotypes. These results identify hepatic glycogen as a key regulator of endurance capacity in mice, an effect that may be exerted through the maintenance of blood glucose levels.


Asunto(s)
Glucemia/metabolismo , Tolerancia al Ejercicio/fisiología , Ácidos Grasos no Esterificados/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Glucógeno Hepático/metabolismo , Músculo Esquelético/metabolismo , Animales , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
Brain ; 144(8): 2349-2360, 2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-33822008

RESUMEN

The hallmark of Lafora disease, a fatal neurodegenerative disorder, is the accumulation of intracellular glycogen aggregates called Lafora bodies. Until recently, it was widely believed that brain Lafora bodies were present exclusively in neurons and thus that Lafora disease pathology derived from their accumulation in this cell population. However, recent evidence indicates that Lafora bodies are also present in astrocytes. To define the role of astrocytic Lafora bodies in Lafora disease pathology, we deleted glycogen synthase specifically from astrocytes in a mouse model of the disease (malinKO). Strikingly, blocking glycogen synthesis in astrocytes-thus impeding Lafora bodies accumulation in this cell type-prevented the increase in neurodegeneration markers, autophagy impairment, and metabolic changes characteristic of the malinKO model. Conversely, mice that over-accumulate glycogen in astrocytes showed an increase in these markers. These results unveil the deleterious consequences of the deregulation of glycogen metabolism in astrocytes and change the perspective that Lafora disease is caused solely by alterations in neurons.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Glucógeno/metabolismo , Enfermedad de Lafora/metabolismo , Degeneración Nerviosa/metabolismo , Animales , Astrocitos/patología , Encéfalo/patología , Modelos Animales de Enfermedad , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Enfermedad de Lafora/genética , Enfermedad de Lafora/patología , Ratones , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
8.
J Biol Chem ; 296: 100498, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33667544

RESUMEN

Hepatic glycogen metabolism is impaired in diabetes. We previously demonstrated that strategies to increase liver glycogen content in a high-fat-diet mouse model of obesity and insulin resistance led to a reduction in food intake and ameliorated obesity and glucose tolerance. These effects were accompanied by a decrease in insulin levels, but whether this decrease contributed to the phenotype observed in this animal was unclear. Here we sought to evaluate this aspect directly, by examining the long-term effects of increasing liver glycogen in an animal model of insulin-deficient and monogenic diabetes, namely the Akita mouse, which is characterized by reduced insulin production. We crossed Akita mice with animals overexpressing protein targeting to glycogen (PTG) in the liver to generate Akita mice with increased liver glycogen content (Akita-PTGOE). Akita-PTGOE animals showed lower glycemia, lower food intake, and decreased water consumption and urine output compared with Akita mice. Furthermore, Akita-PTGOE mice showed a restoration of the hepatic energy state and a normalization of gluconeogenesis and glycolysis back to nondiabetic levels. Moreover, hepatic lipogenesis, which is reduced in Akita mice, was reverted in Akita-PTGOE animals. These results demonstrate that strategies to increase liver glycogen content lead to the long-term reduction of the diabetic phenotype, independently of circulating insulin.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Glucógeno Hepático/metabolismo , Animales , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Gluconeogénesis , Glucólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo
9.
FEBS Lett ; 594(11): 1698-1710, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32159852

RESUMEN

Glycogen shortage during fasting coincides with dramatic changes in hepatic adenine nucleotide levels. The aim of this work was to study the relevance of liver glycogen in the regulation of the hepatic energy state during food deprivation. To this end, we examined the response of mice with sustained increased liver glycogen content to prolonged fasting. In order to increase hepatic glycogen content, we generated mice that overexpress protein targeting to glycogen (PTG) in the liver (PTGOE  mice). Control and PTGOE  mice were fed ad libitum or fasted for 36 h. Upon fasting, PTGOE  mice retained significant hepatic glycogen stores and maintained hepatic energy status. Furthermore, we show that liver glycogen controls insulin sensitivity, gluconeogenesis, lipid metabolism, and ketogenesis upon nutrient deprivation.


Asunto(s)
Metabolismo Energético , Ayuno/metabolismo , Glucógeno/metabolismo , Hígado/metabolismo , Tejido Adiposo , Animales , Peso Corporal , Privación de Alimentos , Gluconeogénesis , Insulina , Resistencia a la Insulina , Cetonas/metabolismo , Metabolismo de los Lípidos , Ratones , Músculo Esquelético/metabolismo , Tamaño de los Órganos
10.
Front Cell Neurosci ; 13: 374, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31456667

RESUMEN

Since brain glycogen is stored mainly in astrocytes, the role of this polysaccharide in neurons has been largely overlooked. To study the existence and relevance of an active neuronal glycogen metabolism in vivo, we generated a mouse model lacking glycogen synthase specifically in the Camk2a-expressing postnatal forebrain pyramidal neurons (GYS1Camk2a-KO), which include the prefrontal cortex and the CA3 and CA1 cell layers of the hippocampus. The latter are involved in memory and learning processes and participate in the hippocampal CA3-CA1 synapse, the function of which can be analyzed electrophysiologically. Long-term potentiation evoked in the hippocampal CA3-CA1 synapse was decreased in alert behaving GYS1Camk2a-KO mice. They also showed a significant deficiency in the acquisition of an instrumental learning task - a type of associative learning involving prefrontal and hippocampal circuits. Interestingly, GYS1Camk2a-KO animals did not show the greater susceptibility to hippocampal seizures and myoclonus observed in animals completely depleted of glycogen in the whole CNS. These results unequivocally demonstrate the presence of an active glycogen metabolism in neurons in vivo and reveal a key role of neuronal glycogen in the proper acquisition of new motor and cognitive abilities, and in the changes in synaptic strength underlying such acquisition.

11.
Eur J Nutr ; 57(8): 2963-2974, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29127477

RESUMEN

PURPOSE: The utilization of long-chain polyunsaturated fatty acids (LCPUFA) by the fetus may exceed its capacity to synthesize them from essential fatty acids, so they have to come from the mother. Since adipose tissue lipolytic activity is greatly accelerated under fasting conditions during late pregnancy, the aim was to determine how 24 h fasting in late pregnant rats given diets with different fatty acid compositions affects maternal and fetal tissue fatty acid profiles. METHODS: Pregnant Sprague-Dawley rats were given isoenergetic diets containing 10% palm-, sunflower-, olive- or fish-oil. Half the rats were fasted from day 19 of pregnancy and all were studied on day 20. Triacylglycerols (TAG), glycerol and non-esterified fatty acids (NEFA) were analyzed by enzymatic methods and fatty acid profiles were analyzed by gas chromatography. RESULTS: Fasting caused increments in maternal plasma NEFA, glycerol and TAG, indicating increased adipose tissue lipolytic activity. Maternal adipose fatty acid profiles paralleled the respective diets and, with the exception of animals on the olive oil diet, maternal fasting increased the plasma concentration of most fatty acids. This maintains the availability of LCPUFA to the fetus during brain development. CONCLUSIONS: The results show the major role played by maternal adipose tissue in the storage of dietary fatty acids during pregnancy, thus ensuring adequate availability of LCPUFA to the fetus during late pregnancy, even when food supply is restricted.


Asunto(s)
Tejido Adiposo/química , Ácidos Grasos/química , Feto/química , Fenómenos Fisiologicos Nutricionales Maternos , Animales , Dieta , Grasas de la Dieta/administración & dosificación , Ayuno , Ácidos Grasos no Esterificados/química , Femenino , Aceites de Pescado/administración & dosificación , Lipólisis , Intercambio Materno-Fetal , Aceite de Oliva/administración & dosificación , Embarazo , Ratas , Ratas Sprague-Dawley , Aceite de Girasol/administración & dosificación , Triglicéridos/sangre
12.
Cell Metab ; 26(1): 256-266.e4, 2017 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-28683291

RESUMEN

Glycogenin is considered essential for glycogen synthesis, as it acts as a primer for the initiation of the polysaccharide chain. Against expectations, glycogenin-deficient mice (Gyg KO) accumulate high amounts of glycogen in striated muscle. Furthermore, this glycogen contains no covalently bound protein, thereby demonstrating that a protein primer is not strictly necessary for the synthesis of the polysaccharide in vivo. Strikingly, in spite of the higher glycogen content, Gyg KO mice showed lower resting energy expenditure and less resistance than control animals when subjected to endurance exercise. These observations can be attributed to a switch of oxidative myofibers toward glycolytic metabolism. Mice overexpressing glycogen synthase in the muscle showed similar alterations, thus indicating that this switch is caused by the excess of glycogen. These results may explain the muscular defects of GSD XV patients, who lack glycogenin-1 and show high glycogen accumulation in muscle.


Asunto(s)
Glucosiltransferasas/metabolismo , Glucógeno/metabolismo , Glicoproteínas/metabolismo , Músculo Esquelético/fisiología , Animales , Metabolismo Energético , Glucosiltransferasas/genética , Glucógeno Sintasa/metabolismo , Glicoproteínas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/metabolismo , Consumo de Oxígeno
13.
Diabetologia ; 60(6): 1076-1083, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28299379

RESUMEN

AIMS/HYPOTHESIS: Liver glycogen plays a key role in regulating food intake and blood glucose. Mice that accumulate large amounts of this polysaccharide in the liver are protected from high-fat diet (HFD)-induced obesity by reduced food intake. Furthermore, these animals show reversal of the glucose intolerance and hyperinsulinaemia caused by the HFD. The aim of this study was to examine the involvement of the hepatic branch of the vagus nerve in regulating food intake and glucose homeostasis in this model. METHODS: We performed hepatic branch vagotomy (HBV) or a sham operation on mice overexpressing protein targeting to glycogen (Ptg OE). Starting 1 week after surgery, mice were fed an HFD for 10 weeks. RESULTS: HBV did not alter liver glycogen or ATP levels, thereby indicating that this procedure does not interfere with hepatic energy balance. However, HBV reversed the effect of glycogen accumulation on food intake. In wild-type mice, HBV led to a significant reduction in body weight without a change in food intake. Consistent with their body weight reduction, these animals had decreased fat deposition, adipocyte size, and insulin and leptin levels, together with increased energy expenditure. Ptg OE mice showed an increase in energy expenditure and glucose oxidation, and these differences were abolished by HBV. Moreover, Ptg OE mice showed an improvement in HFD-induced glucose intolerance, which was suppressed by HBV. CONCLUSIONS/INTERPRETATION: Our results demonstrate that the regulation of food intake and glucose homeostasis by liver glycogen is dependent on the hepatic branch of the vagus nerve.


Asunto(s)
Glucemia/fisiología , Ingestión de Alimentos/fisiología , Glucógeno Hepático/metabolismo , Nervio Vago/metabolismo , Nervio Vago/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Homeostasis , Hígado/metabolismo , Ratones , Obesidad/etiología , Obesidad/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Reacción en Cadena de la Polimerasa
14.
Am J Physiol Endocrinol Metab ; 310(6): E440-51, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26714846

RESUMEN

Neuregulin (NRG) is an EGF-related growth factor that binds to the tyrosine kinase receptors ErbB3 and ErbB4, thus inducing tissue development and muscle glucose utilization during contraction. Here, we analyzed whether NRG has systemic effects regulating glycemia in control and type 2 diabetic rats. To this end, recombinant NRG (rNRG) was injected into Zucker diabetic fatty (ZDF) rats and their respective lean littermates 15 min before a glucose tolerance test (GTT) was performed. rNRG enhanced glucose tolerance without promoting the activation of the insulin receptor (IR) or insulin receptor substrates (IRS) in muscle and liver. However, in control rats, rNRG induced the phosphorylation of protein kinase B (PKB) and glycogen synthase kinase-3 (GSK-3) in liver but not in muscle. In liver, rNRG increased ErbB3 tyrosine phosphorylation and its binding to phosphatidylinositol 3-kinase (PI3K), thus indicating that rNRG activates the ErbB3/PI3K/PKB signaling pathway. rNRG increased glycogen content in liver but not in muscle. rNRG also increased the content of fructose-2,6-bisphosphate (Fru-2,6-P2), an activator of hepatic glycolysis, and lactate in liver but not in muscle. Increases in lactate were abrogated by wortmannin, a PI3K inhibitor, in incubated hepatocytes. The liver of ZDF rats showed a reduced content of ErbB3 receptors, entailing a minor stimulation of the rNRG-induced PKB/GSK-3 cascade and resulting in unaltered hepatic glycogen content. Nonetheless, rNRG increased hepatic Fru-2,6-P2 and augmented lactate both in liver and in plasma of diabetic rats. As a whole, rNRG improved response to the GTT in both control and diabetic rats by enhancing hepatic glucose utilization.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Hígado/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Neurregulinas/farmacología , Animales , Glucemia/metabolismo , Estudios de Casos y Controles , Fructosadifosfatos/metabolismo , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Insulina , Proteínas Sustrato del Receptor de Insulina/efectos de los fármacos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ácido Láctico/metabolismo , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinasa/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Zucker , Receptor ErbB-3/efectos de los fármacos , Receptor ErbB-3/metabolismo , Receptor de Insulina/efectos de los fármacos , Receptor de Insulina/metabolismo
15.
Am J Physiol Endocrinol Metab ; 310(5): E367-77, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26714850

RESUMEN

To investigate the biodisponibility of placental transfer of fatty acids, rats pregnant for 20 days were given tracer amounts of [(14)C]palmitic (PA), oleic (OA), linoleic (LA), α-linolenic (LNA), or docosahexaenoic acid (DHA) orally and euthanized at 0.5, 1.0, 2.0, or 8.0 h thereafter. Maternal plasma radioactivity in lipids initially increased only to decline at later times. Most of the label appeared first as triacylglycerols (TAG); later, the proportion in phospholipids (PhL) increased. The percentage of label in placental lipids was also always highest shortly after administration and declined later; again, PhL increased with time. Fetal plasma radioactivity increased with time, with its highest value at 8.0 h after DHA or LNA administration. DHA initially appeared primarily in the nonesterified fatty acids (NEFA) and PA, OA, LA, and LNA as TAG followed by NEFA; in all cases, there was an increase in PhL at later times. Measurement of fatty acid concentrations allowed calculation of specific (radio)activities, and the ratio (fetal/maternal) of these in the plasmas gave an index of placental transfer activity, which was LNA > LA > DHA = OA > PA. It is proposed that a considerable proportion of most fatty acids transferred through the placenta are released into the fetal circulation in the form of TAG.


Asunto(s)
Ácidos Docosahexaenoicos/farmacocinética , Feto/metabolismo , Ácido Linoleico/farmacocinética , Ácido Oléico/farmacocinética , Ácido Palmítico/farmacocinética , Fosfolípidos/metabolismo , Placenta/metabolismo , Triglicéridos/metabolismo , Ácido alfa-Linolénico/farmacocinética , Animales , Radioisótopos de Carbono , Ácidos Docosahexaenoicos/metabolismo , Ácidos Grasos/metabolismo , Ácidos Grasos/farmacocinética , Femenino , Ácido Linoleico/metabolismo , Ácido Oléico/metabolismo , Ácido Palmítico/metabolismo , Embarazo , Ratas , Ácido alfa-Linolénico/metabolismo
16.
Diabetes ; 64(3): 796-807, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25277398

RESUMEN

We generated mice that overexpress protein targeting to glycogen (PTG) in the liver (PTG(OE)), which results in an increase in liver glycogen. When fed a high-fat diet (HFD), these animals reduced their food intake. The resulting effect was a lower body weight, decreased fat mass, and reduced leptin levels. Furthermore, PTG overexpression reversed the glucose intolerance and hyperinsulinemia caused by the HFD and protected against HFD-induced hepatic steatosis. Of note, when fed an HFD, PTG(OE) mice did not show the decrease in hepatic ATP content observed in control animals and had lower expression of neuropeptide Y and higher expression of proopiomelanocortin in the hypothalamus. Additionally, after an overnight fast, PTG(OE) animals presented high liver glycogen content, lower liver triacylglycerol content, and lower serum concentrations of fatty acids and ß-hydroxybutyrate than control mice, regardless of whether they were fed an HFD or a standard diet. In conclusion, liver glycogen accumulation caused a reduced food intake, protected against the deleterious effects of an HFD, and diminished the metabolic impact of fasting. Therefore, we propose that hepatic glycogen content be considered a potential target for the pharmacological manipulation of diabetes and obesity.


Asunto(s)
Ingestión de Alimentos/fisiología , Glucógeno Hepático/metabolismo , Obesidad/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Dieta Alta en Grasa , Leptina/sangre , Hígado/metabolismo , Glucógeno Hepático/fisiología , Ratones , Obesidad/sangre , Obesidad/prevención & control
17.
Diabetes ; 62(12): 4070-82, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23990365

RESUMEN

The liver responds to an increase in blood glucose levels in the postprandial state by uptake of glucose and conversion to glycogen. Liver glycogen synthase (GYS2), a key enzyme in glycogen synthesis, is controlled by a complex interplay between the allosteric activator glucose-6-phosphate (G6P) and reversible phosphorylation through glycogen synthase kinase-3 and the glycogen-associated form of protein phosphatase 1. Here, we initially performed mutagenesis analysis and identified a key residue (Arg(582)) required for activation of GYS2 by G6P. We then used GYS2 Arg(582)Ala knockin (+/R582A) mice in which G6P-mediated GYS2 activation had been profoundly impaired (60-70%), while sparing regulation through reversible phosphorylation. R582A mutant-expressing hepatocytes showed significantly reduced glycogen synthesis with glucose and insulin or glucokinase activator, which resulted in channeling glucose/G6P toward glycolysis and lipid synthesis. GYS2(+/R582A) mice were modestly glucose intolerant and displayed significantly reduced glycogen accumulation with feeding or glucose load in vivo. These data show that G6P-mediated activation of GYS2 plays a key role in controlling glycogen synthesis and hepatic glucose-G6P flux control and thus whole-body glucose homeostasis.


Asunto(s)
Glucosa-6-Fosfato/metabolismo , Glucógeno Sintasa/metabolismo , Hepatocitos/metabolismo , Glucógeno Hepático/biosíntesis , Hígado/metabolismo , Animales , Glucemia/metabolismo , Glucosa/farmacología , Glucógeno Sintasa/genética , Hepatocitos/efectos de los fármacos , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Insulina/farmacología , Hígado/efectos de los fármacos , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Fosforilación
18.
Am J Physiol Endocrinol Metab ; 298(4): E742-50, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20028964

RESUMEN

Neuregulin was described initially as a neurotrophic factor involved in the formation of the neuromuscular junction in skeletal muscle. However, in recent years, neuregulin has been reported to be a myokine that exerts relevant effects on myogenesis and the regulation of muscle metabolism. In this new context, the rapid and chronic metabolic effects of neuregulin appear to be related to muscle contraction. Indeed, the effects of neuregulin resemble those of exercise, which are accompanied by an improvement in insulin sensitivity. In this review, we challenge the classical role assigned to neuregulin in muscle and propound the emerging concept of its involvement in the regulation of energetic metabolism and insulin responsiveness.


Asunto(s)
Músculos/fisiología , Neurregulinas/fisiología , Animales , Ejercicio Físico/fisiología , Humanos , Ratones , Ratones Transgénicos , Contracción Muscular/fisiología , Músculos/metabolismo , Neurregulinas/metabolismo , Receptores de Superficie Celular/fisiología
19.
Biochim Biophys Acta ; 1791(12): 1181-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19699314

RESUMEN

Dietary n-3 polyunsaturated fatty acids (PUFA) suppress the secretion of very low density lipoprotein (VLDL) directly when delivered to the liver in chylomicron remnants (CMR). The role of sterol regulatory element-binding proteins (SREBPs) and hepatic nuclear factor-4alpha (HNF-4alpha) in the regulation of this effect was investigated. Chylomicron remnant-like particles (CRLPs) containing triacylglycerol (TG) from palm (rich in saturated fatty acids (SFA)) or fish (rich in n-3 PUFA) oil were incubated with cultured rat hepatocytes (24h) and the expression of protein and mRNA for SREBP-1, SREBP-2 and HNF-4alpha, and levels of mRNA for their target genes were determined. SREBP-1 and -2 protein expression in the membrane and nuclear fractions was unaffected by either type of CRLPs. mRNA abundance for SREBP-1c and -2 was also unchanged by CRLP-treatment, as were levels of mRNA for target genes of SREBP-1, including steroyl CoA desaturase, acetyl CoA carboxylase, fatty acid synthase and ATP citrate lyase, and SREBP-2 (3-hydroxy-3-methylglutaryl CoA reductase). In contrast, HNF-4alpha protein and mRNA levels were significantly decreased by CRLPs enriched in n-3 PUFA, but not SFA, and the expression of mRNA for HNF-4alpha target genes, including HNF-1alpha, apolipoprotein B and the microsomal TG transfer protein, was also lowered by n-3 PUFA-, but not SFA-enriched CRLPs. These findings suggest that the direct suppression of VLDL secretion by dietary n-3 PUFA delivered to the liver in CMR is mediated via decreased expression of HNF-4alpha.


Asunto(s)
Remanentes de Quilomicrones/farmacología , Ácidos Grasos Omega-3/farmacología , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Lipoproteínas VLDL/metabolismo , Animales , Apolipoproteínas E/análisis , Células Cultivadas , Aceites de Pescado/química , Aceites de Pescado/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/genética , Masculino , Aceite de Palma , Aceites de Plantas/química , Aceites de Plantas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo
20.
Metabolism ; 58(2): 186-95, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19154951

RESUMEN

The influence of dietary fats carried in chylomicron remnants on the hepatic secretion of very low-density lipoprotein (VLDL) was investigated using chylomicron remnant-like particles (CRLPs) and cultured rat hepatocytes as the experimental model. Chylomicron remnant-like particles containing triacylglycerol (TG) from palm, olive, or corn (enriched in saturated, monounsaturated, or n-6 polyunsaturated fatty acids) oil, respectively, were incubated with cultured hepatocytes for 5 hours. The medium was then removed and replaced with medium without CRLPs; and the secretion of TG, cholesterol, and apolipoprotein B48 during the following 16 hours was determined. Secretion of TG into the d less than 1.050-g/mL fraction containing VLDL was unaffected by olive CRLPs, but was significantly increased in cells exposed to palm or corn CRLPs in comparison with both olive CRLPs and control incubations without CRLPs. Secretion of apolipoprotein B48, however, was not changed by any of the CRLP types. Apolipoprotein B messenger RNA levels were decreased by olive and corn CRLPs, and 3-hydroxy-3-methylglutaryl coenzyme A reductase messenger RNA abundance was increased by palm CRLPs; but expression of other genes involved in the regulation of VLDL secretion was unaffected. These findings demonstrate that CRLPs enriched in saturated fatty acids or n-6 polyunsaturated fatty acids increase the secretion of TG in VLDL, possibly because of the secretion of larger particles, whereas those enriched in monounsaturated fatty acids have no effect. Thus, different dietary fats have differential effects on VLDL secretion directly when delivered to the liver in chylomicron remnants.


Asunto(s)
Remanentes de Quilomicrones/farmacología , Grasas de la Dieta/farmacología , Ácidos Grasos/farmacología , Hepatocitos/metabolismo , Lipoproteínas VLDL/metabolismo , Animales , Apolipoproteínas B/metabolismo , Células Cultivadas , Aceite de Maíz/farmacología , Ácidos Grasos Omega-6/farmacología , Ácidos Grasos Insaturados/farmacología , Glicerol-3-Fosfato O-Aciltransferasa/genética , Hepatocitos/citología , Hidroximetilglutaril-CoA Reductasas/genética , Masculino , Aceite de Oliva , Aceite de Palma , Aceites de Plantas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Esterol O-Aciltransferasa/genética , Triglicéridos/farmacología , Esterol O-Aciltransferasa 2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA