Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Physiol Endocrinol Metab ; 326(1): E14-E28, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37938177

RESUMEN

Regular exercise elicits adaptations in glucose and lipid metabolism that allow the body to meet energy demands of subsequent exercise bouts more effectively and mitigate metabolic diseases including fatty liver. Energy discharged during the acute exercise bouts that comprise exercise training may be a catalyst for liver adaptations. During acute exercise, liver glycogenolysis and gluconeogenesis are accelerated to supply glucose to working muscle. Lower liver energy state imposed by gluconeogenesis and related pathways activates AMP-activated protein kinase (AMPK), which conserves ATP partly by promoting lipid oxidation. This study tested the hypothesis that AMPK is necessary for liver glucose and lipid adaptations to training. Liver-specific AMPKα1α2 knockout (AMPKα1α2fl/fl+AlbCre) mice and littermate controls (AMPKα1α2fl/fl) completed sedentary and exercise training protocols. Liver nutrient fluxes were quantified at rest or during acute exercise following training. Liver metabolites and molecular regulators of metabolism were assessed. Training increased liver glycogen in AMPKα1α2fl/fl mice, but not in AMPKα1α2fl/fl+AlbCre mice. The inability to increase glycogen led to lower glycogenolysis, glucose production, and circulating glucose during acute exercise in trained AMPKα1α2fl/fl+AlbCre mice. Deletion of AMPKα1α2 attenuated training-induced declines in liver diacylglycerides. In particular, training lowered the concentration of unsaturated and elongated fatty acids comprising diacylglycerides in AMPKα1α2fl/fl mice, but not in AMPKα1α2fl/fl+AlbCre mice. Training increased liver triacylglycerides and the desaturation and elongation of fatty acids in triacylglycerides of AMPKα1α2fl/fl+AlbCre mice. These lipid responses were independent of differences in tricarboxylic acid cycle fluxes. In conclusion, AMPK is required for liver training adaptations that are critical to glucose and lipid metabolism.NEW & NOTEWORTHY This study shows that the energy sensor and transducer, AMP-activated protein kinase (AMPK), is necessary for an exercise training-induced: 1) increase in liver glycogen that is necessary for accelerated glycogenolysis during exercise, 2) decrease in liver glycerolipids independent of tricarboxylic acid (TCA) cycle flux, and 3) decline in the desaturation and elongation of fatty acids comprising liver diacylglycerides. The mechanisms defined in these studies have implications for use of regular exercise or AMPK-activators in patients with fatty liver.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Hígado Graso , Humanos , Animales , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Glucógeno Hepático , Hígado/metabolismo , Glucosa/metabolismo , Hígado Graso/metabolismo , Ácidos Grasos/metabolismo
2.
Am J Physiol Endocrinol Metab ; 326(4): E428-E442, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38324258

RESUMEN

Glucagon rapidly and profoundly stimulates hepatic glucose production (HGP), but for reasons that are unclear, this effect normally wanes after a few hours, despite sustained plasma glucagon levels. This study characterized the time course of glucagon-mediated molecular events and their relevance to metabolic flux in the livers of conscious dogs. Glucagon was either infused into the hepato-portal vein at a sixfold basal rate in the presence of somatostatin and basal insulin, or it was maintained at a basal level in control studies. In one control group, glucose remained at basal, whereas in the other, glucose was infused to match the hyperglycemia that occurred in the hyperglucagonemic group. Elevated glucagon caused a rapid (30 min) and largely sustained increase in hepatic cAMP over 4 h, a continued elevation in glucose-6-phosphate (G6P), and activation and deactivation of glycogen phosphorylase and synthase activities, respectively. Net hepatic glycogenolysis increased rapidly, peaking at 15 min due to activation of the cAMP/PKA pathway, then slowly returned to baseline over the next 3 h in line with allosteric inhibition by glucose and G6P. Glucagon's stimulatory effect on HGP was sustained relative to the hyperglycemic control group due to continued PKA activation. Hepatic gluconeogenic flux did not increase due to the lack of glucagon's effect on substrate supply to the liver. Global gene expression profiling highlighted glucagon-regulated activation of genes involved in cellular respiration, metabolic processes, and signaling, as well as downregulation of genes involved in extracellular matrix assembly and development.NEW & NOTEWORTHY Glucagon rapidly stimulates hepatic glucose production, but these effects are transient. This study links the molecular and metabolic flux changes that occur in the liver over time in response to a rise in glucagon, demonstrating the strength of the dog as a translational model to couple findings in small animals and humans. In addition, this study clarifies why the rapid effects of glucagon on liver glycogen metabolism are not sustained.


Asunto(s)
Glucagón , Insulina , Humanos , Perros , Animales , Glucagón/metabolismo , Insulina/metabolismo , Transcriptoma , Glucosa/metabolismo , Hígado/metabolismo , Gluconeogénesis/genética , Glucemia/metabolismo
3.
J Biol Chem ; 293(30): 11944-11954, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-29891549

RESUMEN

Glycine N-methyltransferase (GNMT) is the most abundant liver methyltransferase regulating the availability of the biological methyl donor, S-adenosylmethionine (SAM). Moreover, GNMT has been identified to be down-regulated in hepatocellular carcinoma (HCC). Despite its role in regulating SAM levels and association of its down-regulation with liver tumorigenesis, the impact of reduced GNMT on metabolic reprogramming before the manifestation of HCC has not been investigated in detail. Herein, we used 2H/13C metabolic flux analysis in conscious, unrestrained mice to test the hypothesis that the absence of GNMT causes metabolic reprogramming. GNMT-null (KO) mice displayed a reduction in blood glucose that was associated with a decline in both hepatic glycogenolysis and gluconeogenesis. The reduced gluconeogenesis was due to a decrease in liver gluconeogenic precursors, citric acid cycle fluxes, and anaplerosis and cataplerosis. A concurrent elevation in both hepatic SAM and metabolites of SAM utilization pathways was observed in the KO mice. Specifically, the increase in metabolites of SAM utilization pathways indicated that hepatic polyamine synthesis and catabolism, transsulfuration, and de novo lipogenesis pathways were increased in the KO mice. Of note, these pathways utilize substrates that could otherwise be used for gluconeogenesis. Also, this metabolic reprogramming occurs before the well-documented appearance of HCC in GNMT-null mice. Together, these results indicate that GNMT deletion promotes a metabolic shift whereby nutrients are channeled away from glucose formation toward pathways that utilize the elevated SAM.


Asunto(s)
Carbono/metabolismo , Eliminación de Gen , Gluconeogénesis , Glicina N-Metiltransferasa/genética , Metionina/metabolismo , Animales , Ciclo del Ácido Cítrico , Metabolismo Energético , Hígado Graso/genética , Hígado Graso/metabolismo , Glucosa/metabolismo , Glicina N-Metiltransferasa/metabolismo , Hígado/metabolismo , Masculino , Análisis de Flujos Metabólicos , Ratones , Ratones Noqueados , S-Adenosilmetionina/metabolismo
4.
J Biol Chem ; 292(49): 20125-20140, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29038293

RESUMEN

Pathologies including diabetes and conditions such as exercise place an unusual demand on liver energy metabolism, and this demand induces a state of energy discharge. Hepatic AMP-activated protein kinase (AMPK) has been proposed to inhibit anabolic processes such as gluconeogenesis in response to cellular energy stress. However, both AMPK activation and glucose release from the liver are increased during exercise. Here, we sought to test the role of hepatic AMPK in the regulation of in vivo glucose-producing and citric acid cycle-related fluxes during an acute bout of muscular work. We used 2H/13C metabolic flux analysis to quantify intermediary metabolism fluxes in both sedentary and treadmill-running mice. Additionally, liver-specific AMPK α1 and α2 subunit KO and WT mice were utilized. Exercise caused an increase in endogenous glucose production, glycogenolysis, and gluconeogenesis from phosphoenolpyruvate. Citric acid cycle fluxes, pyruvate cycling, anaplerosis, and cataplerosis were also elevated during this exercise. Sedentary nutrient fluxes in the postabsorptive state were comparable for the WT and KO mice. However, the increment in the endogenous rate of glucose appearance during exercise was blunted in the KO mice because of a diminished glycogenolytic flux. This lower rate of glycogenolysis was associated with lower hepatic glycogen content before the onset of exercise and prompted a reduction in arterial glucose during exercise. These results indicate that liver AMPKα1α2 is required for maintaining glucose homeostasis during an acute bout of exercise.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucogenólisis , Hígado/enzimología , Condicionamiento Físico Animal , Proteínas Quinasas Activadas por AMP/deficiencia , Animales , Metabolismo Energético , Gluconeogénesis , Glucosa/metabolismo , Homeostasis , Marcaje Isotópico , Ratones , Ratones Noqueados
5.
Am J Physiol Endocrinol Metab ; 313(3): E263-E272, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28536182

RESUMEN

The postprandial state is characterized by a storage of nutrients in the liver, muscle, and adipose tissue for later utilization. In the case of a protein-rich meal, amino acids (AA) stimulate glucagon secretion by the α-cell. The aim of the present study was to determine the impact of the rise in glucagon on AA metabolism, particularly in the liver. We used a conscious catheterized dog model to recreate a postprandial condition using a pancreatic clamp. Portal infusions of glucose, AA, and insulin were used to achieve postprandial levels, while portal glucagon infusion was either maintained at the basal level or increased by three-fold. The high glucagon infusion reduced the increase in arterial AA concentrations compared with the basal glucagon level (-23%, P < 0.05). In the presence of high glucagon, liver AA metabolism shifted toward a more catabolic state with less protein synthesis (-36%) and increased urea production (+52%). Net hepatic glucose uptake was reduced modestly (-35%), and AA were preferentially used in gluconeogenesis, leading to lower glycogen synthesis (-54%). The phosphorylation of AMPK was increased by the high glucagon infusion (+40%), and this could be responsible for increasing the expression of genes related to pathways producing energy and lowering those involved in energy consumption. In conclusion, the rise in glucagon associated with a protein-rich meal promotes a catabolic utilization of AA in the liver, thereby, opposing the storage of AA in proteins.


Asunto(s)
Aminoácidos/efectos de los fármacos , Glucemia/efectos de los fármacos , Glucagón/farmacología , Hormonas/farmacología , Hígado/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Aminoácidos/metabolismo , Aminoácidos/farmacología , Animales , Glucemia/metabolismo , Perros , Gluconeogénesis/efectos de los fármacos , Glucosa/metabolismo , Glucosa/farmacología , Hipoglucemiantes/farmacología , Infusiones Intravenosas , Insulina/farmacología , Hígado/metabolismo , Fosforilación/efectos de los fármacos , Vena Porta , Periodo Posprandial , Proteínas/efectos de los fármacos , Proteínas/metabolismo , Urea/metabolismo
6.
Am J Physiol Endocrinol Metab ; 313(3): E273-E283, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28512154

RESUMEN

The contribution of hormone-independent counterregulatory signals in defense of insulin-induced hypoglycemia was determined in adrenalectomized, overnight-fasted conscious dogs receiving hepatic portal vein insulin infusions at a rate 20-fold basal. Either euglycemia was maintained (group 1) or hypoglycemia (≈45 mg/dl) was allowed to occur. There were three hypoglycemic groups: one in which hepatic autoregulation against hypoglycemia occurred in the absence of sympathetic nervous system input (group 2), one in which autoregulation occurred in the presence of norepinephrine (NE) signaling to fat and muscle (group 3), and one in which autoregulation occurred in the presence of NE signaling to fat, muscle, and liver (group 4). Average net hepatic glucose balance (NHGB) during the last hour for groups 1-4 was -0.7 ± 0.1, 0.3 ± 0.1 (P < 0.01 vs. group 1), 0.7 ± 0.1 (P = 0.01 vs. group 2), and 0.8 ± 0.1 (P = 0.7 vs. group 3) mg·kg-1·min-1, respectively. Hypoglycemia per se (group 2) increased NHGB by causing an inhibition of net hepatic glycogen synthesis. NE signaling to fat and muscle (group 3) increased NHGB further by mobilizing gluconeogenic precursors resulting in a rise in gluconeogenesis. Lowering glucose per se decreased nonhepatic glucose uptake by 8.9 mg·kg-1·min-1, and the addition of increased neural efferent signaling to muscle and fat blocked glucose uptake further by 3.2 mg·kg-1·min-1 The addition of increased neural efferent input to liver did not affect NHGB or nonhepatic glucose uptake significantly. In conclusion, even in the absence of increases in counterregulatory hormones, the body can defend itself against hypoglycemia using glucose autoregulation and increased neural efferent signaling, both of which stimulate hepatic glucose production and limit glucose utilization.


Asunto(s)
Glucemia/efectos de los fármacos , Hipoglucemia/metabolismo , Hipoglucemiantes/farmacología , Insulina/farmacología , Hígado/efectos de los fármacos , Tejido Adiposo/metabolismo , Adrenalectomía , Animales , Glucemia/metabolismo , Perros , Gluconeogénesis/efectos de los fármacos , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Homeostasis , Hipoglucemia/inducido químicamente , Infusiones Intravenosas , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Músculo Esquelético/metabolismo , Norepinefrina/metabolismo , Vena Porta , Sistema Nervioso Simpático
7.
J Biol Chem ; 289(9): 5950-9, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24403081

RESUMEN

Metabolic stress, as well as several antidiabetic agents, increases hepatic nucleotide monophosphate (NMP) levels, activates AMP-activated protein kinase (AMPK), and suppresses glucose production. We tested the necessity of hepatic AMPK for the in vivo effects of an acute elevation in NMP on metabolism. 5-Aminoimidazole-4-carboxamide 1-ß-D-ribofuranoside (AICAR; 8 mg·kg(-1)·min(-1))-euglycemic clamps were performed to elicit an increase in NMP in wild type (α1α2(lox/lox)) and liver-specific AMPK knock-out mice (α1α2(lox/lox) + Albcre) in the presence of fixed glucose. Glucose kinetics were equivalent in 5-h fasted α1α2(lox/lox) and α1α2(lox/lox) + Albcre mice. AMPK was not required for AICAR-mediated suppression of glucose production and increased glucose disappearance. These results demonstrate that AMPK is unnecessary for normal 5-h fasting glucose kinetics and AICAR-mediated inhibition of glucose production. Moreover, plasma fatty acids and triglycerides also decreased independently of hepatic AMPK during AICAR administration. Although the glucoregulatory effects of AICAR were shown to be independent of AMPK, these studies provide in vivo support for the AMPK energy sensor paradigm. AICAR reduced hepatic energy charge by ∼20% in α1α2(lox/lox), which was exacerbated by ∼2-fold in α1α2(lox/lox) + Albcre. This corresponded to a ∼6-fold rise in AMP/ATP in α1α2(lox/lox) + Albcre. Consistent with the effects on adenine nucleotides, maximal mitochondrial respiration was ∼30% lower in α1α2(lox/lox) + Albcre than α1α2(lox/lox) livers. Mitochondrial oxidative phosphorylation efficiency was reduced by 25%. In summary, these results demonstrate that the NMP capacity to inhibit glucose production in vivo is independent of liver AMPK. In contrast, AMPK promotes mitochondrial function and protects against a more precipitous fall in ATP during AICAR administration.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Metabolismo Energético , Glucosa/biosíntesis , Hipoglucemiantes/farmacología , Hígado/metabolismo , Ribonucleótidos/farmacología , Proteínas Quinasas Activadas por AMP/genética , Aminoimidazol Carboxamida/farmacología , Animales , Ácidos Grasos/sangre , Glucosa/genética , Hígado/citología , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Triglicéridos/sangre
8.
Am J Physiol Endocrinol Metab ; 306(11): E1225-38, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24714398

RESUMEN

A loss of glucose effectiveness to suppress hepatic glucose production as well as increase hepatic glucose uptake and storage as glycogen is associated with a defective increase in glucose phosphorylation catalyzed by glucokinase (GK) in Zucker diabetic fatty (ZDF) rats. We extended these observations by investigating the role of persistent hyperglycemia (glucotoxicity) in the development of impaired hepatic GK activity in ZDF rats. We measured expression and localization of GK and GK regulatory protein (GKRP), translocation of GK, and hepatic glucose flux in response to a gastric mixed meal load (MMT) and hyperglycemic hyperinsulinemic clamp after 1 or 6 wk of treatment with the sodium-glucose transporter 2 inhibitor (canaglifrozin) that was used to correct the persistent hyperglycemia of ZDF rats. Defective augmentation of glucose phosphorylation in response to a rise in plasma glucose in ZDF rats was associated with the coresidency of GKRP with GK in the cytoplasm in the midstage of diabetes, which was followed by a decrease in GK protein levels due to impaired posttranscriptional processing in the late stage of diabetes. Correcting hyperglycemia from the middle diabetic stage normalized the rate of glucose phosphorylation by maintaining GK protein levels, restoring normal nuclear residency of GK and GKRP under basal conditions and normalizing translocation of GK from the nucleus to the cytoplasm, with GKRP remaining in the nucleus in response to a rise in plasma glucose. This improved the liver's metabolic ability to respond to hyperglycemic hyperinsulinemia. Glucotoxicity is responsible for loss of glucose effectiveness and is associated with altered GK regulation in the ZDF rat.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucoquinasa/metabolismo , Glucosa/toxicidad , Hígado/enzimología , Obesidad/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Canagliflozina , Diabetes Mellitus Tipo 2/complicaciones , Ingestión de Alimentos/efectos de los fármacos , Glucagón/metabolismo , Glucosa/biosíntesis , Técnica de Clampeo de la Glucosa , Glucósidos/farmacología , Hiperglucemia/metabolismo , Hiperglucemia/patología , Hiperinsulinismo/metabolismo , Inmunohistoquímica , Hígado/metabolismo , Masculino , Obesidad/complicaciones , Tamaño de los Órganos/efectos de los fármacos , Consumo de Oxígeno , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Zucker , Transportador 2 de Sodio-Glucosa , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Tiofenos/farmacología
9.
Am J Physiol Endocrinol Metab ; 303(10): E1202-11, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23011060

RESUMEN

Portal vein glucose delivery (the portal glucose signal) stimulates glucose uptake and glycogen storage by the liver, whereas portal amino acid (AA) delivery (the portal AA signal) induces an increase in protein synthesis by the liver. During a meal, both signals coexist and may interact. In this study, we compared the protein synthesis rates in the liver and muscle in response to portal or peripheral glucose infusion during intraportal infusion of a complete AA mixture. Dogs were surgically prepared with hepatic sampling catheters and flow probes. After a 42-h fast, they underwent a 3-h hyperinsulinemic (4× basal) hyperglucagonemic (3× basal) hyperglycemic (≈160 mg/dl) hyperaminoacidemic (hepatic load 1.5× basal; delivered intraportally) clamp (postprandial conditions). Glucose was infused either via a peripheral (PeG; n = 7) or the portal vein (PoG; n = 8). Protein synthesis was assessed with a primed, continuous [(14)C]leucine infusion. Net hepatic glucose uptake was stimulated by portal glucose infusion (+1 mg·kg(-1)·min(-1), P < 0.05) as expected, but hepatic fractional AA extraction and hepatic protein synthesis did not differ between groups. There was a lower arterial AA concentration in the PoG group (-19%, P < 0.05) and a significant stimulation (+30%) of muscle protein synthesis associated with increased expression of LAT1 and ASCT2 AA transporters and p70S6 phosphorylation. Concomitant portal glucose and AA delivery enhances skeletal muscle protein synthesis compared with peripheral glucose and portal AA delivery. These data suggest that enteral nutrition support may have an advantage over parenteral nutrition in stimulating muscle protein synthesis.


Asunto(s)
Glucosa/administración & dosificación , Hígado/efectos de los fármacos , Hígado/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Sistema de Transporte de Aminoácidos ASC/metabolismo , Aminoácidos/metabolismo , Animales , Perros , Nutrición Enteral , Glucosa/metabolismo , Glucosa/farmacocinética , Infusiones Intravenosas , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Masculino , Fosforilación , Vena Porta , Periodo Posprandial , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
10.
J Pharmacol Exp Ther ; 337(3): 610-20, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21363927

RESUMEN

The effects of a glycogen phosphorylase inhibitor (GPI) and metformin (MT) on hepatic glucose fluxes (µmol · kg(-1) · min(-1)) in the presence of basal and 4-fold basal levels of plasma glucagon were investigated in 18-h fasted conscious dogs. Compared with the vehicle treatment, GPI infusion suppressed net hepatic glucose output (NHGO) completely (-3.8 ± 1.3 versus 9.9 ± 2.8) despite increased glucose 6-phosphate (G-6-P) neogenesis from gluconeogenic precursors (8.1 ± 1.1 versus 5.5 ± 1.1). MT infusion did not alter those parameters. In response to a 4-fold rise in plasma glucagon levels, in the vehicle group, plasma glucose levels were increased 2-fold, and NHGO was increased (43.9 ± 5.7 at 10 min and 22.7 ± 3.4 at steady state) without altering G-6-P neogenesis (3.7 ± 1.5 and 5.5 ± 0.5, respectively). In the GPI group, there was no increase in NHGO due to decreased glucose-6-phosphatase flux associated with reduced G-6-P concentration. A lower G-6-P concentration was the result of increased net glycogenesis without altering G-6-P neogenesis. In the MT group, the increment in NHGO (22.2 ± 4.4 at 10 min and 12.1 ± 3.6 at steady state) was approximately half of that of the vehicle group. The lesser NHGO was associated with reduced glucose-6-phosphatase flux but a rise in G-6-P concentration and only a small incorporation of plasma glucose into glycogen. In conclusion, the inhibition of glycogen phosphorylase a activity decreases basal and glucagon-induced NHGO via redirecting glucose 6-phosphate flux from glucose toward glycogen, and MT decreases glucagon-induced NHGO by inhibiting glucose-6-phosphatase flux and thereby reducing glycogen breakdown.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Glucógeno Fosforilasa de Forma Hepática/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Glucógeno Hepático/metabolismo , Hígado/efectos de los fármacos , Metformina/farmacología , Animales , Glucemia/metabolismo , Perros , Ayuno , Ácidos Grasos no Esterificados/sangre , Ácidos Grasos no Esterificados/metabolismo , Femenino , Glucagón/sangre , Glucagón/metabolismo , Glucagón/farmacología , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/fisiología , Glucosa-6-Fosfatasa/efectos de los fármacos , Glucosa-6-Fosfatasa/fisiología , Glicerol/sangre , Glicerol/metabolismo , Glucógeno Fosforilasa de Forma Hepática/metabolismo , Hematócrito , Indoles/farmacología , Insulina/sangre , Insulina/metabolismo , Ácido Láctico/sangre , Ácido Láctico/metabolismo , Hígado/metabolismo , Masculino , Fenilbutiratos/farmacología
11.
Am J Physiol Endocrinol Metab ; 299(4): E607-14, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20663988

RESUMEN

Hepatic glucagon action increases in response to accelerated metabolic demands and is associated with increased whole body substrate availability, including circulating lipids. The hypothesis that increases in hepatic glucagon action stimulate AMP-activated protein kinase (AMPK) signaling and peroxisome proliferator-activated receptor-α (PPARα) and fibroblast growth factor 21 (FGF21) expression in a manner modulated by fatty acids was tested in vivo. Wild-type (gcgr(+/+)) and glucagon receptor-null (gcgr(-/-)) littermate mice were studied using an 18-h fast, exercise, and hyperglucagonemic-euglycemic clamps plus or minus increased circulating lipids. Fasting and exercise in gcgr(+/+), but not gcgr(-/-) mice, increased hepatic phosphorylated AMPKα at threonine 172 (p-AMPK(Thr(172))) and PPARα and FGF21 mRNA. Clamp results in gcgr(+/+) mice demonstrate that hyperlipidemia does not independently impact or modify glucagon-stimulated increases in hepatic AMP/ATP, p-AMPK(Thr(172)), or PPARα and FGF21 mRNA. It blunted glucagon-stimulated acetyl-CoA carboxylase phosphorylation, a downstream target of AMPK, and accentuated PPARα and FGF21 expression. All effects were absent in gcgr(-/-) mice. These findings demonstrate that glucagon exerts a critical regulatory role in liver to stimulate pathways linked to lipid metabolism in vivo and shows for the first time that effects of glucagon on PPARα and FGF21 expression are amplified by a physiological increase in circulating lipids.


Asunto(s)
Adenilato Quinasa/metabolismo , Emulsiones Grasas Intravenosas/metabolismo , Factores de Crecimiento de Fibroblastos/biosíntesis , Glucagón/metabolismo , Hígado/metabolismo , PPAR alfa/biosíntesis , Adenilato Quinasa/genética , Animales , Área Bajo la Curva , Glucemia/metabolismo , Catecolaminas/sangre , Ácidos Grasos no Esterificados/sangre , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Técnica de Clampeo de la Glucosa , Insulina/sangre , Hígado/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR alfa/genética , PPAR alfa/metabolismo , Condicionamiento Físico Animal/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Glucagón/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
12.
Nutr Diabetes ; 9(1): 25, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31474750

RESUMEN

BACKGROUND: Aerobic exercise training is known to have beneficial effects on whole-body glucose metabolism in people with type 2 diabetes (T2D). The responses of the liver to such training are less well understood. The purpose of this study was to determine the effect of aerobic exercise training on splanchnic glucose uptake (SGU) and insulin-mediated suppression of endogenous glucose production (EGP) in obese subjects with T2D. METHODS: Participants included 11 obese humans with T2D, who underwent 15 ± 2 weeks of aerobic exercise training (AEX; n = 6) or remained sedentary for 15 ± 1 weeks (SED; n = 5). After an initial screening visit, each subject underwent an oral glucose load clamp and an isoglycemic/two-step (20 and 40 mU/m2/min) hyperinsulinemic clamp (ISO-clamp) to assess SGU and insulin-mediated suppression of EGP, respectively. After the intervention period, both tests were repeated. RESULTS: In AEX, the ability of insulin to suppress EGP was improved during both the low (69 ± 9 and 80 ± 6% suppression; pre-post, respectively; p < 0.05) and high (67 ± 6 and 82 ± 4% suppression, respectively; p < 0.05) insulin infusion periods. Despite markedly improved muscle insulin sensitivity, SGU was reduced in AEX after training (22.9 ± 3.3 and 9.1 ± 6.0 g pre-post in AEX, respectively; p < 0.05). CONCLUSIONS: In obese T2D subjects, exercise training improves whole-body glucose metabolism, in part, by improving insulin-mediated suppression of EGP and enhancing muscle glucose uptake, which occur despite reduced SGU during an oral glucose challenge.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Ejercicio Físico/fisiología , Glucosa/metabolismo , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Adulto , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Persona de Mediana Edad , Obesidad/complicaciones
13.
Diabetes ; 55(9): 2479-90, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16936196

RESUMEN

The effect of restoration of normoglycemia by a novel sodium-dependent glucose transporter inhibitor (T-1095) on impaired hepatic glucose uptake was examined in 14-week-old Zucker diabetic fatty (ZDF) rats. The nontreated group exhibited persistent endogenous glucose production (EGP) despite marked hyperglycemia. Gluconeogenesis and glucose cycling (GC) were responsible for 46 and 51% of glucose-6-phosphatase (G6Pase) flux, respectively. Net incorporation of plasma glucose into hepatic glycogen was negligible. Glucokinase (GK) and its inhibitory protein, GK regulatory protein (GKRP), were colocalized in the cytoplasm of hepatocytes. At day 7 of drug administration, EGP was slightly reduced, but G6Pase flux and GC were markedly lower compared with the nontreated group. In this case, GK and GKRP were colocalized in the nuclei of hepatocytes. When plasma glucose and insulin levels were raised during a clamp, EGP was completely suppressed and GC, glycogen synthesis from plasma glucose, and the fractional contribution of plasma glucose to uridine diphosphoglucose flux were markedly increased. GK, but not GKRP, was translocated from the nucleus to the cytoplasm. Glucotoxicity may result in the blunted response of hepatic glucose flux to elevated plasma glucose and/or insulin associated with impaired regulation of GK by GKRP in ZDF rats.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucoquinasa/metabolismo , Glucosa/metabolismo , Hiperglucemia/fisiopatología , Animales , Glucemia/metabolismo , Carbonatos/farmacología , Proteínas Portadoras/metabolismo , Ácidos Grasos no Esterificados/sangre , Glucagón/sangre , Glucoquinasa/antagonistas & inhibidores , Glucosa-6-Fosfatasa/metabolismo , Glucósidos/farmacología , Glucógeno/metabolismo , Glucógeno Sintasa/metabolismo , Insulina/sangre , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Masculino , Músculo Esquelético/metabolismo , Fosforilasas/metabolismo , Ratas , Ratas Zucker
15.
PLoS One ; 12(1): e0170382, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28107516

RESUMEN

AMPK is an energy sensor that protects cellular energy state by attenuating anabolic and promoting catabolic processes. AMPK signaling is purported to regulate hepatic gluconeogenesis and substrate oxidation; coordination of these processes is vital during nutrient deprivation or pathogenic during overnutrition. Here we directly test hepatic AMPK function in regulating metabolic fluxes that converge to produce glucose and energy in vivo. Flux analysis was applied in mice with a liver-specific deletion of AMPK (L-KO) or floxed control littermates to assess rates of hepatic glucose producing and citric acid cycle (CAC) fluxes. Fluxes were assessed in short and long term fasted mice; the latter condition is a nutrient stressor that increases liver AMP/ATP. The flux circuit connecting anaplerosis with gluconeogenesis from the CAC was unaffected by hepatic AMPK deletion in short and long term fasting. Nevertheless, depletion of hepatic ATP was exacerbated in L-KO mice, corresponding to a relative elevation in citrate synthase flux and accumulation of branched-chain amino acid-related metabolites. L-KO mice also had a physiological reduction in flux from glycogen to G6P. These results demonstrate AMPK is unnecessary for maintaining gluconeogenic flux from the CAC yet is critical for stabilizing liver energy state during nutrient deprivation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Metabolismo Energético , Gluconeogénesis , Hígado/enzimología , Adenosina Trifosfato/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Diabetes ; 54(2): 373-82, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15677495

RESUMEN

AMP-activated protein kinase (AMPK) plays a key role in regulating metabolism, serving as a metabolic master switch. The aim of this study was to assess whether increased concentrations of the AMP analog, 5-aminoimidazole-4-carboxamide-1-beta-D-ribosyl-5-monophosphate, in the liver would create a metabolic response consistent with an increase in whole-body metabolic need. Dogs had sampling (artery, portal vein, hepatic vein) and infusion (vena cava, portal vein) catheters and flow probes (hepatic artery, portal vein) implanted >16 days before a study. Protocols consisted of equilibration (-130 to -30 min), basal (-30 to 0 min), and hyperinsulinemic-euglycemic or -hypoglycemic clamp periods (0-150 min). At t = 0 min, somatostatin was infused and glucagon was replaced in the portal vein at basal rates. An intraportal hyperinsulinemic (2 mU . kg(-1) . min(-1)) infusion was also initiated at this time. Glucose was clamped at hypoglycemic or euglycemic levels in the presence (H-AIC, n = 6; E-AIC, n = 6) or absence (H-SAL, n = 6; E-SAL, n = 6) of a portal venous 5-aminoimidazole-4-carboxamide-ribofuranoside (AICAR) infusion (1 mg . kg(-1) . min(-1)) initiated at t = 60 min. In the presence of intraportal saline, glucose was infused into the vena cava to match glucose levels seen with intraportal AICAR. Glucagon remained fixed at basal levels, whereas insulin rose similarly in all groups. Glucose fell to 50 +/- 2 mg/dl by t = 60 min in hypoglycemic groups and remained at 105 +/- 3 mg/dl in euglycemic groups. Endogenous glucose production (R(a)) was similarly suppressed among groups in the presence of euglycemia or hypoglycemia before t = 60 min and remained suppressed in the H-SAL and E-SAL groups. However, intraportal AICAR infusion stimulated R(a) to increase by 2.5 +/- 1.0 and 3.4 +/- 0.4 mg . kg(-1) . min(-1) in the E-AIC and H-AIC groups, respectively. Arteriovenous measurement of net hepatic glucose output showed similar results. AICAR stimulated hepatic glycogen to decrease by 5 +/- 3 and 19 +/- 5 mg/g tissue (P < 0.05) in the presence of euglycemia and hypoglycemia, respectively. AICAR significantly increased net hepatic lactate output in the presence of hypoglycemia. Thus, intraportal AICAR infusion caused marked stimulation of both hepatic glucose output and net hepatic glycogenolysis, even in the presence of high levels of physiological insulin. This stimulation of glucose output by AICAR was equally marked in the presence of both euglycemia and hypoglycemia. However, hypoglycemia amplified the net hepatic glycogenolytic response to AICAR by approximately fourfold.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Glucemia/metabolismo , Hiperinsulinismo/prevención & control , Ribonucleótidos/farmacología , Aminoimidazol Carboxamida/administración & dosificación , Animales , Glucemia/efectos de los fármacos , Perros , Femenino , Técnica de Clampeo de la Glucosa , Venas Hepáticas , Infusiones Intravenosas , Masculino , Vena Porta , Ribonucleótidos/administración & dosificación
17.
Diabetes ; 54(2): 355-60, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15677492

RESUMEN

The infusion of 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) causes a rise in tissue concentrations of the AMP analog 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranotide (ZMP), which mimics an elevation of cellular AMP levels. The purpose of this work was to determine the effect of raising hepatic ZMP levels on hepatic insulin action in vivo. Dogs had sampling and infusion catheters as well as flow probes implanted 16 days before an experiment. After an 18-h fast, blood glucose was 82 +/- 1 mg/dl and basal net hepatic glucose output 1.5 +/- 0.2 mg . kg(-1) . min(-1). Dogs received portal venous glucose (3.2 mg . kg(-1) . min(-1)), peripheral venous somatostatin, and basal portal venous glucagon infusions from -90 to 60 min. Physiological hyperinsulinemia was established with a portal insulin infusion (1.2 mU . kg(-1) . min(-1)). Peripheral venous glucose infusion was used to clamp arterial blood glucose at 150 mg/dl. Starting at t = 0 min, dogs received portal venous AICAR infusions of 0, 1, or 2 mg . kg(-1) . min(-1). Net hepatic glucose uptake was 2.4 +/- 0.5 mg . kg(-1) . min(-1) (mean of all groups) before t = 0 min. In the absence of AICAR, net hepatic glucose uptake was 1.9 +/- 0.4 mg . kg(-1) . min(-1) at t = 60 min. The lower-dose AICAR infusion caused a complete suppression of net hepatic glucose uptake (-1.0 +/- 1.7 mg . kg(-1) . min(-1) at t = 60 min). The higher AICAR dose resulted in a profound shift in hepatic glucose balance from net uptake to a marked net output (-6.1 +/- 1.9 mg . kg(-1) . min(-1) at t = 60 min), even in the face of hyperglycemia and hyperinsulinemia. These data show that elevations in hepatic ZMP concentrations, induced by portal venous AICAR infusion, cause acute hepatic insulin resistance. These findings have important implications for the targeting of AMP kinase for the treatment of insulin resistance, using AMP analogs.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Resistencia a la Insulina/fisiología , Hígado/fisiología , Ribonucleótidos/farmacología , Aminoimidazol Carboxamida/administración & dosificación , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , AMP Cíclico/metabolismo , Perros , Femenino , Técnica de Clampeo de la Glucosa , Glucólisis/efectos de los fármacos , Hiperinsulinismo/sangre , Infusiones Intravenosas , Hígado/efectos de los fármacos , Masculino , Técnicas de Placa-Clamp , Vena Porta/fisiología , Ribonucleótidos/administración & dosificación
18.
J Clin Invest ; 126(6): 2236-48, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27140398

RESUMEN

Liver glycogen is important for the counterregulation of hypoglycemia and is reduced in individuals with type 1 diabetes (T1D). Here, we examined the effect of varying hepatic glycogen content on the counterregulatory response to low blood sugar in dogs. During the first 4 hours of each study, hepatic glycogen was increased by augmenting hepatic glucose uptake using hyperglycemia and a low-dose intraportal fructose infusion. After hepatic glycogen levels were increased, animals underwent a 2-hour control period with no fructose infusion followed by a 2-hour hyperinsulinemic/hypoglycemic clamp. Compared with control treatment, fructose infusion caused a large increase in liver glycogen that markedly elevated the response of epinephrine and glucagon to a given hypoglycemia and increased net hepatic glucose output (NHGO). Moreover, prior denervation of the liver abolished the improved counterregulatory responses that resulted from increased liver glycogen content. When hepatic glycogen content was lowered, glucagon and NHGO responses to insulin-induced hypoglycemia were reduced. We conclude that there is a liver-brain counterregulatory axis that is responsive to liver glycogen content. It remains to be determined whether the risk of iatrogenic hypoglycemia in T1D humans could be lessened by targeting metabolic pathway(s) associated with hepatic glycogen repletion.


Asunto(s)
Encéfalo/metabolismo , Hipoglucemia/metabolismo , Glucógeno Hepático/metabolismo , Hígado/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Perros , Femenino , Fructosa/administración & dosificación , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Humanos , Hipoglucemia/sangre , Insulina/metabolismo , Ácido Láctico/metabolismo , Metabolismo de los Lípidos , Masculino , Transducción de Señal
19.
Diabetes ; 52(7): 1649-54, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12829628

RESUMEN

Liver-specific PEPCK knockout mice, which are viable despite markedly abnormal lipid metabolism, exhibit mild hyperglycemia in response to fasting. We used isotopic tracer methods, biochemical measurements, and nuclear magnetic resonance spectroscopy to show that in mice lacking hepatic PEPCK, 1) whole-body glucose turnover is only slightly decreased; 2) whole-body gluconeogenesis from phosphoenolpyruvate, but not from glycerol, is moderately decreased; 3) tricarboxylic acid cycle activity is globally increased, even though pyruvate cycling and anaplerosis are decreased; 4) the liver is unable to synthesize glucose from lactate/pyruvate and produces only a minimal amount of glucose; and 5) glycogen synthesis in both the liver and muscle is impaired. Thus, although mice without hepatic PEPCK have markedly impaired hepatic gluconeogenesis, they are able to maintain a near-normal blood glucose concentration while fasting by increasing extrahepatic gluconeogenesis coupled with diminishing whole-body glucose utilization.


Asunto(s)
Glucemia/metabolismo , Hígado/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/deficiencia , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Inanición/fisiopatología , Animales , Glucoquinasa/metabolismo , Glucosa/metabolismo , Glicerol/metabolismo , Glucógeno Sintasa/metabolismo , Homeostasis , Hígado/enzimología , Glucógeno Hepático/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética
20.
Diabetes ; 64(10): 3439-51, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26085570

RESUMEN

Hypoglycemia limits optimal glycemic control in type 1 diabetes mellitus (T1DM), making novel strategies to mitigate it desirable. We hypothesized that portal (Po) vein insulin delivery would lessen hypoglycemia. In the conscious dog, insulin was infused into the hepatic Po vein or a peripheral (Pe) vein at a rate four times of basal. In protocol 1, a full counterregulatory response was allowed, whereas in protocol 2, glucagon was fixed at basal, mimicking the diminished α-cell response to hypoglycemia seen in T1DM. In protocol 1, glucose fell faster with Pe insulin than with Po insulin, reaching 56 ± 3 vs. 70 ± 6 mg/dL (P = 0.04) at 60 min. The change in area under the curve (ΔAUC) for glucagon was similar between Pe and Po, but the peak occurred earlier in Pe. The ΔAUC for epinephrine was greater with Pe than with Po (67 ± 17 vs. 36 ± 14 ng/mL/180 min). In protocol 2, glucose also fell more rapidly than in protocol 1 and fell faster in Pe than in Po, reaching 41 ± 3 vs. 67 ± 2 mg/dL (P < 0.01) by 60 min. Without a rise in glucagon, the epinephrine responses were much larger (ΔAUC of 204 ± 22 for Pe vs. 96 ± 29 ng/mL/180 min for Po). In summary, Pe insulin delivery exacerbates hypoglycemia, particularly in the presence of a diminished glucagon response. Po vein insulin delivery, or strategies that mimic it (i.e., liver-preferential insulin analogs), should therefore lessen hypoglycemia.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hipoglucemia/inducido químicamente , Insulina/administración & dosificación , Insulina/efectos adversos , Administración Intravenosa , Animales , Glucemia/metabolismo , Perros , Glucagón/farmacología , Glucosa/metabolismo , Humanos , Insulina/uso terapéutico , Masculino , Vena Porta , Somatostatina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA