Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Sci Rep ; 9(1): 1861, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755705

RESUMEN

Many members of the G protein-coupled receptor family, including examples with clear therapeutic potential, remain poorly characterised. This often reflects limited availability of suitable tool ligands with which to interrogate receptor function. In the case of GPR84, currently a target for the treatment of idiopathic pulmonary fibrosis, recent times have seen the description of novel orthosteric and allosteric agonists. Using 2-(hexylthiol)pyrimidine-4,6 diol (2-HTP) and di(5,7-difluoro-1H-indole-3-yl)methane (PSB-16671) as exemplars of each class, in cell lines transfected to express either human or mouse GPR84, both ligands acted as effective on-target activators and with high co-operativity in their interactions. This was also the case in lipopolysaccharide-activated model human and mouse immune cell lines. However in mouse bone-marrow-derived neutrophils, where expression of GPR84 is particularly high, the capacity of PSB-16671 but not of 2-HTP to promote G protein activation was predominantly off-target because it was not blocked by an antagonist of GPR84 and was preserved in neutrophils isolated from GPR84 deficient mice. These results illustrate the challenges of attempting to study and define functions of poorly characterised receptors using ligands that have been developed via medicinal chemistry programmes, but where assessed activity has been limited largely to the initially identified target.


Asunto(s)
Receptores Acoplados a Proteínas G/química , Sitio Alostérico , Animales , Sitios de Unión , Células de la Médula Ósea/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Ligandos , Lipopolisacáridos/metabolismo , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Células RAW 264.7 , Receptores Acoplados a Proteínas G/agonistas , Células THP-1
3.
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
4.
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
5.
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
6.
Mol Cell Endocrinol ; 440: 44-56, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27840174

RESUMEN

Inflammation of adipose tissue in obesity is associated with increased IL-1ß, IL-6 and TNF-α secretion and proposed to contribute to insulin resistance. AMP-activated protein kinase (AMPK) regulates nutrient metabolism and is reported to have anti-inflammatory actions in adipose tissue, yet the mechanisms underlying this remain poorly characterised. The effect of AMPK activation on cytokine-stimulated proinflammatory signalling was therefore assessed in cultured adipocytes. AMPK activation inhibited IL-1ß-stimulated CXCL10 secretion, associated with reduced interleukin-1 receptor associated kinase-4 (IRAK4) phosphorylation and downregulated MKK4/JNK and IKK/IκB/NFκB signalling. AMPK activation inhibited TNF-α-stimulated IKK/IκB/NFκB signalling but had no effect on JNK phosphorylation. The JAK/STAT3 pathway was also suppressed by AMPK after IL-6 stimulation and during adipogenesis. Adipose tissue from AMPKα1-/- mice exhibited increased JNK and STAT3 phosphorylation, supporting suppression of these distinct proinflammatory pathways by AMPK in vivo. The inhibition of multiple pro-inflammatory signalling pathways by AMPK may underlie the reported beneficial effects of AMPK activation in adipose tissue.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/enzimología , Adipocitos/patología , Inflamación/enzimología , Inflamación/patología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Animales , Compuestos de Bifenilo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Ratones , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Pironas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Tiofenos/farmacología
7.
Sci Signal ; 9(453): ra109, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27919027

RESUMEN

Adenosine 5'-monophosphate-activated protein kinase (AMPK) is a pivotal regulator of metabolism at cellular and organismal levels. AMPK also suppresses inflammation. We found that pharmacological activation of AMPK rapidly inhibited the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway in various cells. In vitro kinase assays revealed that AMPK directly phosphorylated two residues (Ser515 and Ser518) within the Src homology 2 domain of JAK1. Activation of AMPK enhanced the interaction between JAK1 and 14-3-3 proteins in cultured vascular endothelial cells and fibroblasts, an effect that required the presence of Ser515 and Ser518 and was abolished in cells lacking AMPK catalytic subunits. Mutation of Ser515 and Ser518 abolished AMPK-mediated inhibition of JAK-STAT signaling stimulated by either the sIL-6Rα/IL-6 complex or the expression of a constitutively active V658F-mutant JAK1 in human fibrosarcoma cells. Clinically used AMPK activators metformin and salicylate enhanced the inhibitory phosphorylation of endogenous JAK1 and inhibited STAT3 phosphorylation in primary vascular endothelial cells. Therefore, our findings reveal a mechanism by which JAK1 function and inflammatory signaling may be suppressed in response to metabolic stress and provide a mechanistic rationale for the investigation of AMPK activators in a range of diseases associated with enhanced activation of the JAK-STAT pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Endoteliales/metabolismo , Janus Quinasa 1/metabolismo , Transducción de Señal/fisiología , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Sustitución de Aminoácidos , Animales , Células Endoteliales/citología , Activación Enzimática , Janus Quinasa 1/genética , Ratones , Ratones Noqueados , Mutación Missense , Fosforilación , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
8.
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
9.
Diabetes ; 65(9): 2784-94, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27381369

RESUMEN

Canagliflozin, dapagliflozin, and empagliflozin, all recently approved for treatment of type 2 diabetes, were derived from the natural product phlorizin. They reduce hyperglycemia by inhibiting glucose reuptake by sodium/glucose cotransporter (SGLT) 2 in the kidney, without affecting intestinal glucose uptake by SGLT1. We now report that canagliflozin also activates AMPK, an effect also seen with phloretin (the aglycone breakdown product of phlorizin), but not to any significant extent with dapagliflozin, empagliflozin, or phlorizin. AMPK activation occurred at canagliflozin concentrations measured in human plasma in clinical trials and was caused by inhibition of Complex I of the respiratory chain, leading to increases in cellular AMP or ADP. Although canagliflozin also inhibited cellular glucose uptake independently of SGLT2, this did not account for AMPK activation. Canagliflozin also inhibited lipid synthesis, an effect that was absent in AMPK knockout cells and that required phosphorylation of acetyl-CoA carboxylase (ACC) 1 and/or ACC2 at the AMPK sites. Oral administration of canagliflozin activated AMPK in mouse liver, although not in muscle, adipose tissue, or spleen. Because phosphorylation of ACC by AMPK is known to lower liver lipid content, these data suggest a potential additional benefit of canagliflozin therapy compared with other SGLT2 inhibitors.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Canagliflozina/farmacología , Glucosa/metabolismo , Mitocondrias/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Monofosfato/metabolismo , Animales , Femenino , Células HEK293 , Humanos , Inmunoprecipitación , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Fosforilación , Transportador 2 de Sodio-Glucosa/genética , Inhibidores del Cotransportador de Sodio-Glucosa 2
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...