Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 37(11): e23209, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37779421

RESUMEN

The roles of DGAT1 and DGAT2 in lipid metabolism and insulin responsiveness of human skeletal muscle were studied using cryosections and myotubes prepared from muscle biopsies from control, athlete, and impaired glucose regulation (IGR) cohorts of men. The previously observed increases in intramuscular triacylglycerol (IMTG) in athletes and IGR were shown to be related to an increase in lipid droplet (LD) area in type I fibers in athletes but, conversely, in type II fibers in IGR subjects. Specific inhibition of both diacylglycerol acyltransferase (DGAT) 1 and 2 decreased fatty acid (FA) uptake by myotubes, whereas only DGAT2 inhibition also decreased fatty acid oxidation. Fatty acid uptake in myotubes was negatively correlated with the lactate thresholds of the respective donors. DGAT2 inhibition lowered acetate uptake and oxidation in myotubes from all cohorts whereas DGAT1 inhibition had no effect. A positive correlation between acetate oxidation in myotubes and resting metabolic rate (RMR) from fatty acid oxidation in vivo was observed. Myotubes from athletes and IGR had higher rates of de novo lipogenesis from acetate that were normalized by DGAT2 inhibition. Moreover, DGAT2 inhibition in myotubes also resulted in increased insulin-induced Akt phosphorylation. The differential effects of DGAT1 and DGAT2 inhibition suggest that the specialized role of DGAT2 in esterifying nascent diacylglycerols and de novo synthesized FA is associated with synthesis of a pool of triacylglycerol, which upon hydrolysis results in effectors that promote mitochondrial fatty acid oxidation but decrease insulin signaling in skeletal muscle cells.


Asunto(s)
Diacilglicerol O-Acetiltransferasa , Fibras Musculares Esqueléticas , Masculino , Humanos , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Glucosa/metabolismo , Insulina , Acetatos , Triglicéridos/metabolismo , Ácidos Grasos/metabolismo
2.
Biochem J ; 478(3): 633-646, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33493298

RESUMEN

Activation of AMP-activated protein kinase (AMPK) is considered a valid strategy for the treatment of type 2 diabetes. However, despite the importance of adipose tissue for whole-body energy homeostasis, the effect of AMPK activation in adipocytes has only been studied to a limited extent and mainly with the AMP-mimetic 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR), which has limited specificity. The aim of this study was to evaluate the effect of the allosteric AMPK activators A-769662 and 991 on glucose uptake in adipocytes. For this purpose, primary rat or human adipocytes, and cultured 3T3-L1 adipocytes, were treated with either of the allosteric activators, or AICAR, and basal and insulin-stimulated glucose uptake was assessed. Additionally, the effect of AMPK activators on insulin-stimulated phosphorylation of Akt and Akt substrate of 160 kDa was assessed. Furthermore, primary adipocytes from ADaM site binding drug-resistant AMPKß1 S108A knock-in mice were employed to investigate the specificity of the drugs for the observed effects. Our results show that insulin-stimulated adipocyte glucose uptake was significantly reduced by A-769662 but not 991, yet neither activator had any clear effects on basal or insulin-stimulated Akt/AS160 signaling. The use of AMPKß1 S108A mutant-expressing adipocytes revealed that the observed inhibition of glucose uptake by A-769662 is most likely AMPK-independent, a finding which is supported by the rapid inhibitory effect A-769662 exerts on glucose uptake in 3T3-L1 adipocytes. These data suggest that AMPK activation per se does not inhibit glucose uptake in adipocytes and that the effects of AICAR and A-769662 are AMPK-independent.


Asunto(s)
Adenilato Quinasa/fisiología , Adipocitos/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Glucosa/metabolismo , Pironas/farmacología , Tiofenos/farmacología , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/metabolismo , Sitio Alostérico , Sustitución de Aminoácidos , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Bencimidazoles/farmacología , Benzoatos/farmacología , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Femenino , Técnicas de Sustitución del Gen , Humanos , Insulina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología
3.
Biochem J ; 478(15): 2977-2997, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34259310

RESUMEN

SBI-0206965, originally identified as an inhibitor of the autophagy initiator kinase ULK1, has recently been reported as a more potent and selective AMP-activated protein kinase (AMPK) inhibitor relative to the widely used, but promiscuous inhibitor Compound C/Dorsomorphin. Here, we studied the effects of SBI-0206965 on AMPK signalling and metabolic readouts in multiple cell types, including hepatocytes, skeletal muscle cells and adipocytes. We observed SBI-0206965 dose dependently attenuated AMPK activator (991)-stimulated ACC phosphorylation and inhibition of lipogenesis in hepatocytes. SBI-0206965 (≥25 µM) modestly inhibited AMPK signalling in C2C12 myotubes, but also inhibited insulin signalling, insulin-mediated/AMPK-independent glucose uptake, and AICA-riboside uptake. We performed an extended screen of SBI-0206965 against a panel of 140 human protein kinases in vitro, which showed SBI-0206965 inhibits several kinases, including members of AMPK-related kinases (NUAK1, MARK3/4), equally or more potently than AMPK or ULK1. This screen, together with molecular modelling, revealed that most SBI-0206965-sensitive kinases contain a large gatekeeper residue with a preference for methionine at this position. We observed that mutation of the gatekeeper methionine to a smaller side chain amino acid (threonine) rendered AMPK and ULK1 resistant to SBI-0206965 inhibition. These results demonstrate that although SBI-0206965 has utility for delineating AMPK or ULK1 signalling and cellular functions, the compound potently inhibits several other kinases and critical cellular functions such as glucose and nucleoside uptake. Our study demonstrates a role for the gatekeeper residue as a determinant of the inhibitor sensitivity and inhibitor-resistant mutant forms could be exploited as potential controls to probe specific cellular effects of SBI-0206965.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Benzamidas/farmacología , Pirimidinas/farmacología , Proteínas Recombinantes/metabolismo , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Benzamidas/metabolismo , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Mutación Missense , Unión Proteica/efectos de los fármacos , Multimerización de Proteína , Pirimidinas/metabolismo , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
4.
Clin Sci (Lond) ; 135(13): 1563-1590, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34231841

RESUMEN

Despite obesity and diabetes markedly increasing the risk of developing cardiovascular diseases, the molecular and cellular mechanisms that underlie this association remain poorly characterised. In the last 20 years it has become apparent that chronic, low-grade inflammation in obese adipose tissue may contribute to the risk of developing insulin resistance and type 2 diabetes. Furthermore, increased vascular pro-inflammatory signalling is a key event in the development of cardiovascular diseases. Overnutrition exacerbates pro-inflammatory signalling in vascular and adipose tissues, with several mechanisms proposed to mediate this. In this article, we review the molecular and cellular mechanisms by which nutrients are proposed to regulate pro-inflammatory signalling in adipose and vascular tissues. In addition, we examine the potential therapeutic opportunities that these mechanisms provide for suppression of inappropriate inflammation in obesity and vascular disease.


Asunto(s)
Tejido Adiposo/metabolismo , Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Metabolismo Energético , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Estado Nutricional , Obesidad/metabolismo , Adipoquinas/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/inmunología , Tejido Adiposo/fisiopatología , Animales , Antiinflamatorios/uso terapéutico , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/prevención & control , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/inmunología , Sistema Cardiovascular/fisiopatología , Metabolismo Energético/efectos de los fármacos , Humanos , Hipoglucemiantes/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/fisiopatología , Mediadores de Inflamación/antagonistas & inhibidores , Obesidad/tratamiento farmacológico , Obesidad/inmunología , Obesidad/fisiopatología , Estrés Oxidativo , Transducción de Señal
5.
Biochem J ; 476(12): 1725-1740, 2019 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-31189568

RESUMEN

AMP-activated protein kinase (AMPK) is a heterotrimer of α-catalytic and ß- and γ-regulatory subunits that acts to regulate cellular and whole-body nutrient metabolism. The key role of AMPK in sensing energy status has led to significant interest in AMPK as a therapeutic target for dysfunctional metabolism in type 2 diabetes, insulin resistance and obesity. Despite the actions of AMPK in the liver and skeletal muscle being extensively studied, the role of AMPK in adipose tissue and adipocytes remains less well characterised. Small molecules that selectively influence AMPK heterotrimers containing specific AMPKß subunit isoforms have been developed, including MT47-100, which selectively inhibits complexes containing AMPKß2. AMPKß1 and AMPKß2 are the principal AMPKß subunit isoforms in rodent liver and skeletal muscle, respectively, yet the contribution of specific AMPKß isoforms to adipose tissue function, however, remains largely unknown. This study therefore sought to determine the contribution of AMPKß subunit isoforms to adipocyte biology, focussing on adipogenesis. AMPKß2 was the principal AMPKß isoform in 3T3-L1 adipocytes, isolated rodent adipocytes and human subcutaneous adipose tissue, as assessed by the contribution to total cellular AMPK activity. Down-regulation of AMPKß2 with siRNA inhibited lipid accumulation, cellular adiponectin levels and adiponectin secretion during 3T3-L1 adipogenesis, whereas down-regulation of AMPKß1 had no effect. Incubation of 3T3-L1 cells with MT47-100 selectively inhibited AMPK complexes containing AMPKß2 whilst simultaneously inhibiting cellular lipid accumulation as well as cellular levels and secretion of adiponectin. Taken together, these data indicate that increased expression of AMPKß2 is an important feature of efficient adipogenesis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/enzimología , Adipogénesis , Regulación Enzimológica de la Expresión Génica , Metabolismo de los Lípidos , Regulación hacia Arriba , Células 3T3-L1 , Animales , Humanos , Isoenzimas/metabolismo , Masculino , Ratones , Ratas Sprague-Dawley
6.
Circ Res ; 120(11): 1825-1841, 2017 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-28546359

RESUMEN

The AMP-activated protein kinase (AMPK) is a key regulator of cellular and whole-body energy homeostasis, which acts to restore energy homoeostasis whenever cellular energy charge is depleted. Over the last 2 decades, it has become apparent that AMPK regulates several other cellular functions and has specific roles in cardiovascular tissues, acting to regulate cardiac metabolism and contractile function, as well as promoting anticontractile, anti-inflammatory, and antiatherogenic actions in blood vessels. In this review, we discuss the role of AMPK in the cardiovascular system, including the molecular basis of mutations in AMPK that alter cardiac physiology and the proposed mechanisms by which AMPK regulates vascular function under physiological and pathophysiological conditions.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Enfermedades Cardiovasculares/enzimología , Sistema Cardiovascular/enzimología , Transducción de Señal/fisiología , Animales , Enfermedades Cardiovasculares/patología , Sistema Cardiovascular/patología , Humanos , Remodelación Vascular/fisiología
7.
Pharmacol Res ; 128: 88-100, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29037480

RESUMEN

Exaggerated Janus kinase-signal transducer and activator of transcription (JAK-STAT) signalling is key to the pathogenesis of pro-inflammatory disorders, such as rheumatoid arthritis and cardiovascular diseases. Mutational activation of JAKs is also responsible for several haematological malignancies, including myeloproliferative neoplasms and acute lymphoblastic leukaemia. Accumulating evidence links adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), an energy sensor and regulator of organismal and cellular metabolism, with the suppression of immune and inflammatory processes. Recent studies have shown that activation of AMPK can limit JAK-STAT-dependent signalling pathways via several mechanisms. These novel findings support AMPK activation as a strategy for management of an array of disorders characterised by hyper-activation of the JAK-STAT pathway. This review discusses the pivotal role of JAK-STAT signalling in a range of disorders and how both established clinically used and novel AMPK activators might be used to treat these conditions.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Animales , Humanos , Inflamación/metabolismo , Interleucina-6/metabolismo , Transducción de Señal
8.
Int J Mol Sci ; 19(12)2018 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-30563079

RESUMEN

Protein kinase B (Akt) is a key enzyme in the insulin signalling cascade, required for insulin-stimulated NO production in endothelial cells (ECs). Previous studies have suggested that AMP-activated protein kinase (AMPK) activation stimulates NO synthesis and enhances insulin-stimulated Akt activation, yet these studies have largely used indirect activators of AMPK. The effects of the allosteric AMPK activator A769662 on insulin signalling and endothelial function was therefore examined in cultured human macrovascular ECs. Surprisingly, A769662 inhibited insulin-stimulated NO synthesis and Akt phosphorylation in human ECs from umbilical veins (HUVECs) and aorta (HAECs). In contrast, the AMPK activators compound 991 and AICAR had no substantial inhibitory effect on insulin-stimulated Akt phosphorylation in ECs. Inhibition of AMPK with SBI-0206965 had no effect on the inhibition of insulin-stimulated Akt phosphorylation by A769662, suggesting the inhibitory action of A769662 is AMPK-independent. A769662 decreased IGF1-stimulated Akt phosphorylation yet had no effect on VEGF-stimulated Akt signalling in HUVECs, suggesting that A769662 attenuates early insulin/IGF1 signalling. The effects of A769662 on insulin-stimulated Akt phosphorylation were specific to human ECs, as no effect was observed in the human cancer cell lines HepG2 or HeLa, as well as in mouse embryonic fibroblasts (MEFs). A769662 inhibited insulin-stimulated Erk1/2 phosphorylation in HAECs and MEFs, an effect that was independent of AMPK in MEFs. Therefore, despite being a potent AMPK activator, A769662 has effects unlikely to be mediated by AMPK in human macrovascular ECs that reduce insulin sensitivity and eNOS activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Aorta/enzimología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pironas/farmacología , Tiofenos/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Aorta/citología , Compuestos de Bifenilo , Activación Enzimática/efectos de los fármacos , Células HeLa , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo
9.
Biochem J ; 473(24): 4681-4697, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27784766

RESUMEN

The key metabolic regulator, AMP-activated protein kinase (AMPK), is reported to be down-regulated in metabolic disorders, but the mechanisms are poorly characterised. Recent studies have identified phosphorylation of the AMPKα1/α2 catalytic subunit isoforms at Ser487/491, respectively, as an inhibitory regulation mechanism. Vascular endothelial growth factor (VEGF) stimulates AMPK and protein kinase B (Akt) in cultured human endothelial cells. As Akt has been demonstrated to be an AMPKα1 Ser487 kinase, the effect of VEGF on inhibitory AMPK phosphorylation in cultured primary human endothelial cells was examined. Stimulation of endothelial cells with VEGF rapidly increased AMPKα1 Ser487 phosphorylation in an Akt-independent manner, without altering AMPKα2 Ser491 phosphorylation. In contrast, VEGF-stimulated AMPKα1 Ser487 phosphorylation was sensitive to inhibitors of protein kinase C (PKC) and PKC activation using phorbol esters or overexpression of PKC-stimulated AMPKα1 Ser487 phosphorylation. Purified PKC and Akt both phosphorylated AMPKα1 Ser487 in vitro with similar efficiency. PKC activation was associated with reduced AMPK activity, as inhibition of PKC increased AMPK activity and phorbol esters inhibited AMPK, an effect lost in cells expressing mutant AMPKα1 Ser487Ala. Consistent with a pathophysiological role for this modification, AMPKα1 Ser487 phosphorylation was inversely correlated with insulin sensitivity in human muscle. These data indicate a novel regulatory role of PKC to inhibit AMPKα1 in human cells. As PKC activation is associated with insulin resistance and obesity, PKC may underlie the reduced AMPK activity reported in response to overnutrition in insulin-resistant metabolic and vascular tissues.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteína Quinasa C/metabolismo , Línea Celular , Células HEK293 , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Clin Sci (Lond) ; 124(8): 491-507, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23298225

RESUMEN

AMPK (AMP-activated protein kinase) is a key regulator of cellular and whole-body energy balance. AMPK phosphorylates and regulates many proteins concerned with nutrient metabolism, largely acting to suppress anabolic ATP-consuming pathways while stimulating catabolic ATP-generating pathways. This has led to considerable interest in AMPK as a therapeutic target for the metabolic dysfunction observed in obesity and insulin resistance. The role of AMPK in skeletal muscle and the liver has been extensively studied, such that AMPK has been demonstrated to inhibit synthesis of fatty acids, cholesterol and isoprenoids, hepatic gluconeogenesis and translation while increasing fatty acid oxidation, muscle glucose transport, mitochondrial biogenesis and caloric intake. The role of AMPK in the other principal metabolic and insulin-sensitive tissue, adipose, remains poorly characterized in comparison, yet increasing evidence supports an important role for AMPK in adipose tissue function. Obesity is characterized by hypertrophy of adipocytes and the development of a chronic sub-clinical pro-inflammatory environment in adipose tissue, leading to increased infiltration of immune cells. This combination of dysfunctional hypertrophic adipocytes and a pro-inflammatory environment contributes to insulin resistance and the development of Type 2 diabetes. Exciting recent studies indicate that AMPK may not only influence metabolism in adipocytes, but also act to suppress this pro-inflammatory environment, such that targeting AMPK in adipose tissue may be desirable to normalize adipose dysfunction and inflammation. In the present review, we discuss the role of AMPK in adipose tissue, focussing on the regulation of carbohydrate and lipid metabolism, adipogenesis and pro-inflammatory pathways in physiological and pathophysiological conditions.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo/enzimología , Diabetes Mellitus Tipo 2/enzimología , Obesidad/enzimología , Proteínas Quinasas Activadas por AMP/genética , Tejido Adiposo/citología , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Inflamación/enzimología , Inflamación/genética , Obesidad/inmunología , Obesidad/metabolismo
11.
Clin Sci (Lond) ; 124(6): 403-11, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23057846

RESUMEN

The aim of the present study was to determine whether the endothelial dysfunction associated with CAD (coronary artery disease) and T2D (Type 2 diabetes mellitus) is concomitant with elevated mtROS (mitochondrial reactive oxygen species) production in the endothelium and establish if this, in turn, regulates the activity of endothelial AMPK (AMP-activated protein kinase). We investigated endothelial function, mtROS production and AMPK activation in saphenous veins from patients with advanced CAD. Endothelium-dependent vasodilation was impaired in patients with CAD and T2D relative to those with CAD alone. Levels of mitochondrial H(2)O(2) and activity of AMPK were significantly elevated in primary HSVECs (human saphenous vein endothelial cells) from patients with CAD and T2D compared with those from patients with CAD alone. Incubation with the mitochondria-targeted antioxidant, MitoQ(10) significantly reduced AMPK activity in HSVECs from patients with CAD and T2D but not in cells from patients with CAD alone. Elevated mtROS production in the endothelium of patients with CAD and T2D increases AMPK activation, supporting a role for the kinase in defence against oxidative stress. Further investigation is required to determine whether pharmacological activators of AMPK will prove beneficial in the attenuation of endothelial dysfunction in patients with CAD and T2D.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Endotelio Vascular/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Anciano , Enfermedad de la Arteria Coronaria/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Vasodilatación/fisiología
12.
Cell Signal ; 107: 110676, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37028778

RESUMEN

Asymmetric dimethylarginine (ADMA) is generated through the irreversible methylation of arginine residues. It is an independent risk factor for cardiovascular disease, currently thought to be due to its ability to act as a competitive inhibitor of the nitric oxide (NO) synthase enzymes. Plasma ADMA concentrations increase with obesity and fall following weight loss; however, it is unknown whether they play an active role in adipose pathology. Here, we demonstrate that ADMA drives lipid accumulation through a newly identified NO-independent pathway via the amino-acid sensitive calcium-sensing receptor (CaSR). ADMA treatment of 3T3-L1 and HepG2 cells upregulates a suite of lipogenic genes with an associated increase in triglyceride content. Pharmacological activation of CaSR mimics ADMA while negative modulation of CaSR inhibits ADMA driven lipid accumulation. Further investigation using CaSR overexpressing HEK293 cells demonstrated that ADMA potentiates CaSR signalling via Gq intracellular Ca2+ mobilisation. This study identifies a signalling mechanism for ADMA as an endogenous ligand of the G protein-coupled receptor CaSR that potentially contributes to the impact of ADMA in cardiometabolic disease.


Asunto(s)
Arginina , Receptores Sensibles al Calcio , Humanos , Células HEK293 , Arginina/metabolismo , Óxido Nítrico Sintasa/metabolismo , Lípidos
13.
Biochem Biophys Res Commun ; 412(4): 747-51, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21871446

RESUMEN

Insulin stimulates endothelial NO synthesis, at least in part mediated by phosphorylation and activation of endothelial NO synthase at Ser1177 and Ser615 by Akt. We have previously demonstrated that insulin-stimulated NO synthesis is inhibited under high culture glucose conditions, without altering Ca(2+)-stimulated NO synthesis or insulin-stimulated phosphorylation of eNOS. This indicates that stimulation of endothelial NO synthase phosphorylation may be required, yet not sufficient, for insulin-stimulated nitric oxide synthesis. In the current study we investigated the role of supply of the eNOS substrate, L-arginine as a candidate parallel mechanism underlying insulin-stimulated NO synthesis in cultured human aortic endothelial cells. Insulin rapidly stimulated L-arginine transport, an effect abrogated by incubation with inhibitors of phosphatidylinositol-3'-kinase or infection with adenoviruses expressing a dominant negative mutant Akt. Furthermore, supplementation of endothelial cells with extracellular L-arginine enhanced insulin-stimulated NO synthesis, an effect reversed by co-incubation with the L-arginine transport inhibitor, L-lysine. Basal L-arginine transport was significantly increased under high glucose culture conditions, yet insulin-stimulated L-arginine transport remained unaltered. The increase in L-arginine transport elicited by high glucose was independent of the expression of the cationic amino acid transporters, hCAT1 and hCAT2 and not associated with any changes in the activity of ERK1/2, Akt or protein kinase C (PKC). We propose that rapid stimulation of L-arginine transport contributes to insulin-stimulated NO synthesis in human endothelial cells, yet attenuation of this is unlikely to underlie the inhibition of insulin-stimulated NO synthesis under high glucose conditions.


Asunto(s)
Aorta/efectos de los fármacos , Arginina/metabolismo , Endotelio Vascular/efectos de los fármacos , Insulina/farmacología , Óxido Nítrico/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Aorta/metabolismo , Transporte Biológico/efectos de los fármacos , Línea Celular , Endotelio Vascular/metabolismo , Humanos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética
14.
Biochem J ; 426(1): 85-90, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19925457

RESUMEN

Insulin stimulates endothelial NO (nitric oxide) synthesis via PKB (protein kinase B)/Akt-mediated phosphorylation and activation of eNOS (endothelial NO synthase) at Ser-1177. In previous studies, we have demonstrated that stimulation of eNOS phosphorylation at Ser-1177 may be required, yet is not sufficient for insulin-stimulated NO synthesis. We therefore investigated the role of phosphorylation of eNOS at alternative sites to Ser-1177 as candidate parallel mechanisms contributing to insulin-stimulated NO synthesis. Stimulation of human aortic endothelial cells with insulin rapidly stimulated phosphorylation of both Ser-615 and Ser-1177 on eNOS, whereas phosphorylation of Ser-114, Thr-495 and Ser-633 was unaffected. Insulin-stimulated Ser-615 phosphorylation was abrogated by incubation with the PI3K (phosphoinositide 3-kinase) inhibitor wortmannin, infection with adenoviruses expressing a dominant-negative mutant PKB/Akt or pre-incubation with TNFalpha (tumour necrosis factor alpha), but was unaffected by high culture glucose concentrations. Mutation of Ser-615 to alanine reduced insulin-stimulated NO synthesis, whereas mutation of Ser-615 to aspartic acid increased NO production by NOS in which Ser-1177 had been mutated to an aspartic acid residue. We propose that the rapid PKB-mediated stimulation of phosphorylation of Ser-615 contributes to insulin-stimulated NO synthesis.


Asunto(s)
Hipoglucemiantes/farmacología , Insulina/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/biosíntesis , Fosforilación/efectos de los fármacos , Serina/metabolismo , Western Blotting , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Óxido Nítrico Sintasa de Tipo III/genética
15.
Cell Signal ; 76: 109807, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33038517

RESUMEN

AMP-activated protein kinase (AMPK) is the downstream component of a protein kinase cascade that is a key regulator of energy balance at both the cellular and whole-body level. AMPK acts to stimulate ATP production and reduce ATP consumption when cellular ATP levels fall, thereby normalizing energy balance. Given the central role of AMPK in cellular carbohydrate and lipid metabolism, AMPK activation has been proposed to be a therapeutic target for conditions associated with dysfunctional nutrient metabolism including obesity, type 2 diabetes, hepatic steatosis, cardiovascular diseases and cancer. One way by which increased ATP production can be achieved is by increasing the supply of nutrient substrates. In the 1990s, AMPK activation was demonstrated to stimulate glucose uptake in striated muscle, thereby improving substrate supply for ATP production. Subsequently AMPK activation was postulated to underlie the increase in glucose uptake that occurs during muscle contraction. More recently, however, several lines of evidence have demonstrated that AMPK activation is unlikely to be required for contraction-mediated glucose uptake. Furthermore, despite the importance of AMPK in cellular and whole-body metabolism, far fewer studies have investigated either the role of AMPK in glucose uptake by non-muscle tissues or whether AMPK regulates the uptake of fatty acids. In the present review, we discuss the role of AMPK in nutrient uptake by tissues, focusing on glucose uptake out with muscle and fatty acid uptake.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Glucosa/metabolismo , Metabolismo de los Lípidos , Enfermedades Metabólicas/metabolismo , Animales , Humanos
16.
Arterioscler Thromb Vasc Biol ; 28(12): 2255-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18802013

RESUMEN

OBJECTIVE: Proatherosclerotic adhesion of leukocytes to the endothelium is attenuated by NO. As AMP-activated protein kinase (AMPK) regulates endothelial NO synthesis, we investigated the modulation of adhesion to cultured human aortic endothelial cells (HAECs) by AMPK. METHODS AND RESULTS: HAECs incubated with the AMPK activator, AICAR, or expressing constitutively active AMPK demonstrated reduced TNFalpha-stimulated adhesion of promonocytic U-937 cells. Rapid inhibition of TNFalpha-stimulated U-937 cell adhesion by AICAR was NO-dependent, associated with unaltered cell surface adhesion molecule expression, and reduced MCP-1 secretion by HAECs. In contrast, inhibition of TNFalpha-stimulated U-937 cell adhesion by prolonged AMPK activation was NO-independent and associated with reduced cell surface adhesion molecule expression. CONCLUSIONS: AMPK activation in HAECs inhibits TNFalpha-stimulated leukocyte adhesion by a rapid NO-dependent mechanism associated with reduced MCP-1 secretion and a late NO-independent mechanism whereby adhesion molecule expression, in particular E-selectin, is suppressed.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Monocitos/efectos de los fármacos , Monocitos/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Células Cultivadas , Selectina E/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ribonucleótidos/farmacología , Células U937 , Molécula 1 de Adhesión Celular Vascular/metabolismo
17.
Methods Mol Biol ; 1998: 239-250, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31250307

RESUMEN

The fission yeast Schizosaccharomyces pombe, an ascomycete fungus, is an established model organism for studying eukaryotic molecular and cellular events such as the cell cycle due to its powerful genetics, a sequenced genome, and the ease of molecular manipulation (Wood et al., Nature 415:871-880, 2002; Hoffman et al., Genetics 201:403-423, 2015). This chapter describes genetic and cytological methods to study endosomal sorting complex required for transport (ESCRT) function during the cell cycle in fission yeast. These include tetrad analysis to allow the creation of double mutants to test for genetic interactions by synthetic phenotype characterization, such as cellular growth and the analysis of division septa by calcofluor-white staining.


Asunto(s)
Ciclo Celular , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Microscopía Intravital/métodos , Proteínas de Schizosaccharomyces pombe/fisiología , Schizosaccharomyces/fisiología , Bencenosulfonatos/química , Técnicas de Cultivo de Célula/métodos , Colorantes Fluorescentes/química , Técnicas de Genotipaje/métodos , Microscopía Fluorescente/métodos , Mutación , Coloración y Etiquetado/métodos
18.
Methods Mol Biol ; 1732: 307-319, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29480484

RESUMEN

In addition to the well-characterized role of AMPK in the regulation of nutrient metabolism, it is increasingly clear that AMPK activation has multiple actions on inflammatory signalling. Here we describe methods to identify effects of AMPK activity on pro-inflammatory signalling, specifically (1) the nuclear localization of the key inflammatory mediators nuclear factor-κB (NFκB) and phosphorylated c-Jun N-terminal kinase (JNK), (2) preparation of conditioned medium to analyze the secretion of cytokines/chemokines, and (3) the pro-inflammatory adhesion of leukocytes to cultured cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/etiología , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Células Cultivadas , Medios de Cultivo Condicionados , Activadores de Enzimas/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Leucocitos Mononucleares , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
19.
Front Physiol ; 9: 51, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29479319

RESUMEN

Background and aim: Perivascular adipose tissue (PVAT) positively regulates vascular function through production of factors such as adiponectin but this effect is attenuated in obesity. The enzyme AMP-activated protein kinase (AMPK) is present in PVAT and is implicated in mediating the vascular effects of adiponectin. In this study, we investigated the effect of an obesogenic high fat diet (HFD) on aortic PVAT and whether any changes involved AMPK. Methods: Wild type Sv129 (WT) and AMPKα1 knockout (KO) mice aged 8 weeks were fed normal diet (ND) or HFD (42% kcal fat) for 12 weeks. Adiponectin production by PVAT was assessed by ELISA and AMPK expression studied using immunoblotting. Macrophages in PVAT were identified using immunohistochemistry and markers of M1 and M2 macrophage subtypes evaluated using real time-qPCR. Vascular responses were measured in endothelium-denuded aortic rings with or without attached PVAT. Carotid wire injury was performed and PVAT inflammation studied 7 days later. Key results: Aortic PVAT from KO and WT mice was morphologically indistinct but KO PVAT had more infiltrating macrophages. HFD caused an increased infiltration of macrophages in WT mice with increased expression of the M1 macrophage markers Nos2 and Il1b and the M2 marker Chil3. In WT mice, HFD reduced the anticontractile effect of PVAT as well as reducing adiponectin secretion and AMPK phosphorylation. PVAT from KO mice on ND had significantly reduced adiponectin secretion and no anticontractile effect and feeding HFD did not alter this. Wire injury induced macrophage infiltration of PVAT but did not cause further infiltration in KO mice. Conclusions: High-fat diet causes an inflammatory infiltrate, reduced AMPK phosphorylation and attenuates the anticontractile effect of murine aortic PVAT. Mice lacking AMPKα1 phenocopy many of the changes in wild-type aortic PVAT after HFD, suggesting that AMPK may protect the vessel against deleterious changes in response to HFD.

20.
Sci Rep ; 8(1): 5276, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29588466

RESUMEN

Recent clinical trials of the hypoglycaemic sodium-glucose co-transporter-2 (SGLT2) inhibitors, which inhibit renal glucose reabsorption, have reported beneficial cardiovascular outcomes. Whether SGLT2 inhibitors directly affect cardiovascular tissues, however, remains unclear. We have previously reported that the SGLT2 inhibitor canagliflozin activates AMP-activated protein kinase (AMPK) in immortalised cell lines and murine hepatocytes. As AMPK has anti-inflammatory actions in vascular cells, we examined whether SGLT2 inhibitors attenuated inflammatory signalling in cultured human endothelial cells. Incubation with clinically-relevant concentrations of canagliflozin, but not empagliflozin or dapagliflozin activated AMPK and inhibited IL-1ß-stimulated adhesion of pro-monocytic U937 cells and secretion of IL-6 and monocyte chemoattractant protein-1 (MCP-1). Inhibition of MCP-1 secretion was attenuated by expression of dominant-negative AMPK and was mimicked by the direct AMPK activator, A769662. Stimulation of cells with either canagliflozin or A769662 had no effect on IL-1ß-stimulated cell surface levels of adhesion molecules or nuclear factor-κB signalling. Despite these identical effects of canagliflozin and A769662, IL-1ß-stimulated IL-6/MCP-1 mRNA was inhibited by canagliflozin, but not A769662, whereas IL-1ß-stimulated c-jun N-terminal kinase phosphorylation was inhibited by A769662, but not canagliflozin. These data indicate that clinically-relevant canagliflozin concentrations directly inhibit endothelial pro-inflammatory chemokine/cytokine secretion by AMPK-dependent and -independent mechanisms without affecting early IL-1ß signalling.


Asunto(s)
Proteínas Quinasas Activadas por AMP/inmunología , Antiinflamatorios/farmacología , Canagliflozina/farmacología , Citocinas/inmunología , Células Endoteliales/efectos de los fármacos , Interleucina-1beta/antagonistas & inhibidores , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Animales , Células Cultivadas , Quimiocinas/inmunología , Células Endoteliales/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-1beta/inmunología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA