Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Diabetologia ; 63(8): 1603-1615, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32472192

RESUMEN

AIMS/HYPOTHESIS: Chronic stimulation of ß2-adrenoceptors, opposite to acute treatment, was reported to reduce blood glucose levels, as well as to improve glucose and insulin tolerance in rodent models of diabetes by essentially unknown mechanisms. We recently described a novel pathway that mediates glucose uptake in skeletal muscle cells via stimulation of ß2-adrenoceptors. In the current study we further explored the potential therapeutic relevance of ß2-adrenoceptor stimulation to improve glucose homeostasis and the mechanisms responsible for the effect. METHODS: C57Bl/6N mice with diet-induced obesity were treated both acutely and for up to 42 days with a wide range of clenbuterol dosages and treatment durations. Glucose homeostasis was assessed by glucose tolerance test. We also measured in vivo glucose uptake in skeletal muscle, insulin sensitivity by insulin tolerance test, plasma insulin levels, hepatic lipids and glycogen. RESULTS: Consistent with previous findings, acute clenbuterol administration increased blood glucose and insulin levels. However, already after 4 days of treatment, beneficial effects of clenbuterol were manifested in glucose homeostasis (32% improvement of glucose tolerance after 4 days of treatment, p < 0.01) and these effects persisted up to 42 days of treatment. These favourable metabolic effects could be achieved with doses as low as 0.025 mg kg-1 day-1 (40 times lower than previously studied). Mechanistically, these effects were not due to increased insulin levels, but clenbuterol enhanced glucose uptake in skeletal muscle in vivo both acutely in lean mice (by 64%, p < 0.001) as well as during chronic treatment in diet-induced obese mice (by 74%, p < 0.001). Notably, prolonged treatment with low-dose clenbuterol improved whole-body insulin sensitivity (glucose disposal rate after insulin injection increased up to 1.38 ± 0.31%/min in comparison with 0.15 ± 0.36%/min in control mice, p < 0.05) and drastically reduced hepatic steatosis (by 40%, p < 0.01) and glycogen (by 23%, p < 0.05). CONCLUSIONS/INTERPRETATION: Clenbuterol improved glucose tolerance after 4 days of treatment and these effects were maintained for up to 42 days. Effects were achieved with doses in a clinically relevant microgram range. Mechanistically, prolonged treatment with a low dose of clenbuterol improved glucose homeostasis in insulin resistant mice, most likely by stimulating glucose uptake in skeletal muscle and improving whole-body insulin sensitivity as well as by reducing hepatic lipids and glycogen. We conclude that selective ß2-adrenergic agonists might be an attractive potential treatment for type 2 diabetes. This remains to be confirmed in humans. Graphical abstract.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Clenbuterol/uso terapéutico , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Glucosa/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Animales , Homeostasis/efectos de los fármacos , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo
2.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R666-R677, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30892909

RESUMEN

The type 2 diabetes epidemic makes it important to find insulin-independent ways to improve glucose homeostasis. This study examines the mechanisms activated by a dual ß2-/ß3-adrenoceptor agonist, BRL37344, to increase glucose uptake in skeletal muscle and its effects on glucose homeostasis in vivo. We measured the effect of BRL37344 on glucose uptake, glucose transporter 4 (GLUT4) translocation, cAMP levels, ß2-adrenoceptor desensitization, ß-arrestin recruitment, Akt, AMPK, and mammalian target of rapamycin (mTOR) phosphorylation using L6 skeletal muscle cells as a model. We further tested the ability of BRL37344 to modulate skeletal muscle glucose metabolism in animal models (glucose tolerance tests and in vivo and ex vivo skeletal muscle glucose uptake). In L6 cells, BRL37344 increased GLUT4 translocation and glucose uptake only by activation of ß2-adrenoceptors, with a similar potency and efficacy to that of the nonselective ß-adrenoceptor agonist isoprenaline, despite being a partial agonist with respect to cAMP generation. GLUT4 translocation occurred independently of Akt and AMPK phosphorylation but was dependent on mTORC2. Furthermore, in contrast to isoprenaline, BRL37344 did not promote agonist-mediated desensitization and failed to recruit ß-arrestin1/2 to the ß2-adrenoceptor. In conclusion, BRL37344 improved glucose tolerance and increased glucose uptake into skeletal muscle in vivo and ex vivo through a ß2-adrenoceptor-mediated mechanism independently of Akt. BRL37344 was a partial agonist with respect to cAMP, but a full agonist for glucose uptake, and importantly did not cause classical receptor desensitization or internalization of the receptor.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Etanolaminas/farmacología , Transportador de Glucosa de Tipo 4/metabolismo , Glucosa/metabolismo , Músculo Esquelético/efectos de los fármacos , Mioblastos Esqueléticos/efectos de los fármacos , Receptores Adrenérgicos beta 2/efectos de los fármacos , Animales , Línea Celular , AMP Cíclico/metabolismo , Femenino , Transportador de Glucosa de Tipo 4/genética , Humanos , Cinética , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Transporte de Proteínas , Ratas , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo , Transducción de Señal
3.
Mol Metab ; 85: 101931, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38796310

RESUMEN

OBJECTIVE: Simultaneous activation of ß2- and ß3-adrenoceptors (ARs) improves whole-body metabolism via beneficial effects in skeletal muscle and brown adipose tissue (BAT). Nevertheless, high-efficacy agonists simultaneously targeting these receptors whilst limiting activation of ß1-ARs - and thus inducing cardiovascular complications - are currently non-existent. Therefore, we here developed and evaluated the therapeutic potential of a novel ß2-and ß3-AR, named ATR-127, for the treatment of obesity and its associated metabolic perturbations in preclinical models. METHODS: In the developmental phase, we assessed the impact of ATR-127's on cAMP accumulation in relation to the non-selective ß-AR agonist isoprenaline across various rodent ß-AR subtypes, including neonatal rat cardiomyocytes. Following these experiments, L6 muscle cells were stimulated with ATR-127 to assess the impact on GLUT4-mediated glucose uptake and intramyocellular cAMP accumulation. Additionally, in vitro, and in vivo assessments are conducted to measure ATR-127's effects on BAT glucose uptake and thermogenesis. Finally, diet-induced obese mice were treated with 5 mg/kg ATR-127 for 21 days to investigate the effects on glucose homeostasis, body weight, fat mass, skeletal muscle glucose uptake, BAT thermogenesis and hepatic steatosis. RESULTS: Exposure of L6 muscle cells to ATR-127 robustly enhanced GLUT4-mediated glucose uptake despite low intramyocellular cAMP accumulation. Similarly, ATR-127 markedly increased BAT glucose uptake and thermogenesis both in vitro and in vivo. Prolonged treatment of diet-induced obese mice with ATR-127 dramatically improved glucose homeostasis, an effect accompanied by decreases in body weight and fat mass. These effects were paralleled by an enhanced skeletal muscle glucose uptake, BAT thermogenesis, and improvements in hepatic steatosis. CONCLUSIONS: Our results demonstrate that ATR-127 is a highly effective, novel ß2- and ß3-ARs agonist holding great therapeutic promise for the treatment of obesity and its comorbidities, whilst potentially limiting cardiovascular complications. As such, the therapeutic effects of ATR-127 should be investigated in more detail in clinical studies.


Asunto(s)
Tejido Adiposo Pardo , Ratones Endogámicos C57BL , Músculo Esquelético , Animales , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Masculino , Ratas , Obesidad/metabolismo , Obesidad/tratamiento farmacológico , Hígado Graso/metabolismo , Hígado Graso/tratamiento farmacológico , Termogénesis/efectos de los fármacos , Agonistas Adrenérgicos/farmacología
4.
Pharmacol Res Perspect ; 8(5): e00643, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32813332

RESUMEN

The ß3 -adrenoceptor agonist mirabegron is approved for use for overactive bladder and has been purported to be useful in the treatment of obesity-related metabolic diseases in humans, including those involving disturbances of glucose homeostasis. We investigated the effect of mirabegron on glucose homeostasis with in vitro and in vivo models, focusing on its selectivity at ß-adrenoceptors, ability to cause browning of white adipocytes, and the role of UCP1 in glucose homeostasis. In mouse brown, white, and brite adipocytes, mirabegron-mediated effects were examined on cyclic AMP, UCP1 mRNA, [3 H]-2-deoxyglucose uptake, cellular glycolysis, and O2 consumption. Mirabegron increased cyclic AMP levels, UCP1 mRNA content, glucose uptake, and cellular glycolysis in brown adipocytes, and these effects were either absent or reduced in white adipocytes. In brite adipocytes, mirabegron increased cyclic AMP levels and UCP1 mRNA content resulting in increased UCP1-mediated oxygen consumption, glucose uptake, and cellular glycolysis. The metabolic effects of mirabegron in both brown and brite adipocytes were primarily due to actions at ß3 -adrenoceptors as they were largely absent in adipocytes derived from ß3 -adrenoceptor knockout mice. In vivo, mirabegron increased whole body oxygen consumption, glucose uptake into brown and inguinal white adipose tissue, and improved glucose tolerance, all effects that required the presence of the ß3 -adrenoceptor. Furthermore, in UCP1 knockout mice, the effects of mirabegron on glucose tolerance were attenuated. Thus, mirabegron had effects on cellular metabolism in adipocytes that improved glucose handling in vivo, and were primarily due to actions at the ß3 -adrenoceptor.


Asunto(s)
Acetanilidas/administración & dosificación , Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Agonistas de Receptores Adrenérgicos beta 3/administración & dosificación , Glucólisis/efectos de los fármacos , Tiazoles/administración & dosificación , Proteína Desacopladora 1/genética , Acetanilidas/farmacología , Adenosina Monofosfato/metabolismo , Adipocitos Beige/efectos de los fármacos , Adipocitos Marrones/efectos de los fármacos , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Células CHO , Células Cultivadas , Cricetulus , Desoxiglucosa/metabolismo , Técnicas de Inactivación de Genes , Masculino , Ratones , Oxígeno/metabolismo , Tiazoles/farmacología
5.
J Cell Mol Med ; 13(9B): 3061-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18671761

RESUMEN

Mutations in parkin cause autosomal recessive forms of Parkinson's disease (PD), with an early age of onset and similar pathological phenotype to the idiopathic disease. Parkin has been identified as an E3 ubiquitin ligase that mediates different types of ubiquitination, which has made the search for substrates an intriguing possibility to identify pathological mechanisms linked to PD. In this study, we present PLCgamma1 as a novel substrate for parkin. This association was found in non-transfected human neuroblastoma SH-SY5Y cells as well as in stable cell lines expressing parkin WT and familial mutants R42P and G328E. Analysis of cortical, striatal and nigral human brain homogenates revealed that the interaction between parkin and PLCgamma1 is consistent throughout these regions, suggesting that the interaction is likely to have a physiological relevance for humans. Unlike many of the previously identified substrates, we could also show that the steady-state levels of PLCgamma1 is significantly higher in parkin KO mice and lower in parkin WT human neuroblastoma cells, suggesting that parkin ubiquitination of PLCgamma1 is required for proteasomal degradation. In line with this idea, we show that the ability to ubiquitinate PLCgamma1 in vitro differs significantly between WT and familial mutant parkin. In this study, we demonstrate that parkin interacts with PLCgamma1, affecting PLCgamma1 steady state protein levels in human and murine models with manipulated parkin function and expression levels. This finding could be of relevance for finding novel pathogenic mechanisms leading to PD.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Fosfolipasa C gamma/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Estructura Terciaria de Proteína , Ubiquitina/química
6.
Neurosci Lett ; 436(1): 77-80, 2008 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-18367332

RESUMEN

Presenilin (PS1 and PS2) mutations cause early-onset familial Alzheimer's disease (AD). In addition to affecting beta-amyloid precursor protein (APP) processing and Abeta generation, PSs regulate a number of signaling pathways. We previously showed that PSs regulate both phospholipase C (PLC) and protein kinase C (PKC) alpha and gamma activities. We also reported that PS double knockout mouse embryonic fibroblasts (MEFs) have reduced levels of PKCalpha and enhanced levels of PKCdelta. Here, we determined whether the PS modulation of PLC/PKC has consequences for extracellular regulated kinase (Erk) signaling. Erk has been suggested to be important in AD pathology by modulating APP processing and tau phosphorylation. We found that knocking out PS1 or PS2 alone resulted in increased Erk activity and that this effect could be reversed by the PKCalpha inhibitor Gö6976. We also found that Erk activity following either PLC or PKC stimulation was significantly lower in PS double knockout cells and that treatment with the PKC activator phorbol 12,13-dibutyrate (PdBu) down-regulated total-Erk levels in all cells except PS double knockouts. These results demonstrate that PSs regulate Erk activity through a PKCalpha dependent pathway and that disruption of PLC/PKC signaling in the absence of both PS1 and PS2 results in lower downstream activation of Erk.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Presenilinas/metabolismo , Proteína Quinasa C-alfa/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Immunoblotting , Ratones , Ratones Noqueados , Fosfolipasas de Tipo C/metabolismo
7.
Br J Pharmacol ; 175(21): 4072-4082, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29243229

RESUMEN

The ß3 -adrenoceptor was initially an attractive target for several pharmaceutical companies due to its high expression in rodent adipose tissue, where its activation resulted in decreased adiposity and improved metabolic outputs (such as glucose handling) in animal models of obesity and Type 2 diabetes. However, several drugs acting at the ß3 -adrenoceptor failed in clinical trials. This was thought to be due to their lack of efficacy at the human receptor. Recently, mirabegron, a ß3 -adrenoceptor agonist with human efficacy, was approved in North America, Europe, Japan and Australia for the treatment of overactive bladder syndrome. There are indications that mirabegron may act at other receptors/targets, but whether they have any clinical relevance is relatively unknown. Besides overactive bladder syndrome, mirabegron may have other uses such as in the treatment of heart failure or metabolic disease. This review gives an overview of the off-target effects of mirabegron and its potential use in the treatment of other diseases. LINKED ARTICLES: This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.


Asunto(s)
Acetanilidas/farmacología , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Tiazoles/farmacología , Vejiga Urinaria/efectos de los fármacos , Animales , Humanos , Receptores Adrenérgicos beta 3/metabolismo , Vejiga Urinaria/metabolismo
8.
Cell Signal ; 42: 54-66, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28970184

RESUMEN

Recruitment and activation of brite (or beige) adipocytes has been advocated as a potential avenue for manipulating whole-body energy expenditure. Despite numerous studies illustrating the differences in gene and protein markers between brown, brite and white adipocytes, there is very little information on the adrenergic regulation and function of these brite adipocytes. We have compared the functional (cyclic AMP accumulation, oxygen consumption rates, mitochondrial function, glucose uptake, extracellular acidification rates, calcium influx) profiles of mouse adipocytes cultured from three contrasting depots, namely interscapular brown adipose tissue, and inguinal or epididymal white adipose tissues, following chronic treatment with the peroxisome proliferator-activated receptor γ (PPARγ) agonist rosiglitazone. Prototypical brown adipocytes readily express ß3-adrenoceptors, and ß3-adrenoceptor stimulation increases cyclic AMP accumulation, oxygen consumption rates, mitochondrial function, glucose uptake, and extracellular acidification rates. Treatment of brown adipocytes with rosiglitazone increases uncoupling protein 1 (UCP1) levels, and increases ß3-adrenoceptor mitochondrial function but does not affect glucose uptake responses. In contrast, inguinal white adipocytes only express UCP1 and ß3-adrenoceptors following rosiglitazone treatment, which results in an increase in all ß3-adrenoceptor-mediated functions. The effect of rosiglitazone in epididymal white adipocytes, was much lower compared to inguinal white adipocytes. Rosiglitazone also increased α1-adrenoceptor mediated increases in calcium influx and glucose uptake (but not mitochondrial function) in inguinal and epididymal white adipocytes. In conclusion, the PPARγ agonist rosiglitazone promotes the induction and function of brite adipocytes cultured from inguinal and epididymal white adipose depots.


Asunto(s)
Adipocitos Beige/efectos de los fármacos , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Mitocondrias/efectos de los fármacos , Receptores Adrenérgicos beta 3/genética , Tiazolidinedionas/farmacología , Adipocitos Beige/citología , Adipocitos Beige/metabolismo , Adipocitos Marrones/citología , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Animales , Transporte Biológico , AMP Cíclico/agonistas , AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Masculino , Ratones , Mitocondrias/metabolismo , Especificidad de Órganos , Consumo de Oxígeno/efectos de los fármacos , PPAR gamma/agonistas , PPAR gamma/genética , PPAR gamma/metabolismo , Cultivo Primario de Células , Receptores Adrenérgicos beta 3/metabolismo , Rosiglitazona , Transducción de Señal , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
9.
Physiol Behav ; 92(1-2): 93-7, 2007 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-17568632

RESUMEN

Presenilin proteins, mutated forms of which cause early onset familial Alzheimer's disease, are capable of modulating various cell signal transduction pathways, the most extensively studied of which has been intracellular calcium signalling. Disease causing presenilin mutations can potentiate inositol(1,4,5)trisphosphate (InsP3) mediated endoplasmic reticulum release due to calcium overload in this organelle, as well as attenuate capacitative calcium entry. Our own studies have shown a novel function for presenilins that involves regulation of acetylcholine muscarinic receptor-stimulated phospholipase C upstream of InsP3 regulated calcium release. This article reviews the mechanisms by which presenilins modulate intracellular calcium signalling and the role that deregulated calcium homeostasis could play in the pathogenesis of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Señalización del Calcio/fisiología , Presenilinas/fisiología , Transducción de Señal/fisiología , Animales , Humanos , Fosfolipasas de Tipo C/fisiología
10.
Mol Metab ; 6(6): 611-619, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28580291

RESUMEN

OBJECTIVE: Today, the presence and activity of brown adipose tissue (BAT) in adult humans is generally equated with the induced accumulation of [2-18F]2-fluoro-2-deoxy-d-glucose ([18F]FDG) in adipose tissues, as investigated by positron emission tomography (PET) scanning. In reality, PET-FDG is currently the only method available for in vivo quantification of BAT activity in adult humans. The underlying assumption is that the glucose uptake reflects the thermogenic activity of the tissue. METHODS: To examine this basic assumption, we here followed [18F]FDG uptake by PET and by tissue [3H]-2-deoxy-d-glucose uptake in wildtype and UCP1(-/-) mice, i.e. in mice that do or do not possess the unique thermogenic and calorie-consuming ability of BAT. RESULTS: Unexpectedly, we found that ß3-adrenergically induced (by CL-316,243) glucose uptake was UCP1-independent. Thus, whereas PET-FDG scans adequately reflect glucose uptake, this acute glucose uptake is not secondary to thermogenesis but is governed by an independent cellular signalling, here demonstrated to be mediated via the previously described KU-0063794-sensitive mTOR pathway. CONCLUSIONS: Thus, PET-FDG scans do not exclusively reveal active BAT deposits but rather any tissue possessing an adrenergically-mediated glucose uptake pathway. In contrast, we found that the marked glucose uptake-ameliorating effect of prolonged ß3-adrenergic treatment was UCP1 dependent. Thus, therapeutically, UCP1 activity is required for any anti-diabetic effect of BAT activation.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Fluorodesoxiglucosa F18/farmacocinética , Serina-Treonina Quinasas TOR/metabolismo , Proteína Desacopladora 1/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Animales , Ratones , Ratones Endogámicos C57BL , Proteína Desacopladora 1/genética
11.
Mol Nutr Food Res ; 60(1): 18-42, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26201764

RESUMEN

There are two types of adipose tissue with distinct functions-white adipose tissue stores chemical energy as triglycerides, whereas brown adipose tissue consumes energy and releases heat (thermogenesis) in response to sympathetic nerve activity. In humans, treatments that promote greater brown adipose tissue deposition and/or activity would be highly beneficial in regimes aimed at reducing obesity. Adult humans have restricted populations of prototypical brown adipocytes in the neck and chest areas, but recent advances have established that adipocytes with similar properties, termed "brite" adipocytes, can be recruited in subcutaneous depots thought to be primarily white adipose tissue. These brite adipocytes express the protein machinery required for thermogenesis, but to assess brite adipocytes as viable therapeutic targets we need to understand how to promote conversion of white adipocytes to brite adipocytes and ways to increase optimal energy consumption and thermogenesis in these brite adipocytes. This can be accomplished by pharmacological and nutritional therapies to differing degrees, as reviewed in detail here.


Asunto(s)
Obesidad/dietoterapia , Obesidad/tratamiento farmacológico , Termogénesis/efectos de los fármacos , Termogénesis/fisiología , Adipocitos/citología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/fisiología , Animales , Dieta , Humanos , Modelos Animales , Obesidad/prevención & control
12.
Diabetes ; 63(12): 4115-29, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25008179

RESUMEN

There is an increasing worldwide epidemic of type 2 diabetes that poses major health problems. We have identified a novel physiological system that increases glucose uptake in skeletal muscle but not in white adipocytes. Activation of this system improves glucose tolerance in Goto-Kakizaki rats or mice fed a high-fat diet, which are established models for type 2 diabetes. The pathway involves activation of ß2-adrenoceptors that increase cAMP levels and activate cAMP-dependent protein kinase, which phosphorylates mammalian target of rapamycin complex 2 (mTORC2) at S2481. The active mTORC2 causes translocation of GLUT4 to the plasma membrane and glucose uptake without the involvement of Akt or AS160. Stimulation of glucose uptake into skeletal muscle after activation of the sympathetic nervous system is likely to be of high physiological relevance because mTORC2 activation was observed at the cellular, tissue, and whole-animal level in rodent and human systems. This signaling pathway provides new opportunities for the treatment of type 2 diabetes.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Complejos Multiproteicos/metabolismo , Músculo Esquelético/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Tipo 2/etiología , Dieta Alta en Grasa/efectos adversos , Prueba de Tolerancia a la Glucosa , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal
13.
Neurochem Int ; 60(5): 533-42, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22366649

RESUMEN

Aggresomes are cytoplasmic inclusions which are localized at the microtubule organizing center (MTOC) as a result of induced proteasome inhibition, stress or over-expression of certain proteins. Aggresomes are linked to the pathogenesis of many neurodegenerative diseases. Here we studied whether amyloid precursor protein (APP), a type-I transmembrane glycoprotein, is localized in aggresomes after exposure to stress condition. Using confocal microscopy we found that APP is located in aggresomes and co-localized with vimentin, γ-tubulin, 20S and ubiquitin at the MTOC in response to proteasome dysfunction. An interaction between vimentin and APP was found after proteasome inhibition suggesting that APP is an additional protein constituent of aggresomes. Suppression of the proteasome system in APP-HEK293 cells overexpressing APP or transfected with APP Swedish mutation caused an accumulation of stable, detergent-insoluble forms of APP containing poly-ubiquitinated proteins. In addition, brain homogenates from transgenic mice expressing human APP with the Arctic mutation demonstrated an interaction between APP and the aggresomal-marker vimentin. These data suggest that malfunctioning of the proteasome system caused by mutation or overexpression of pathological or non-pathological proteins may lead to the accumulation of stable aggresomes, perhaps contributing to the neurodegeneration.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores de Proteasoma , Precursor de Proteína beta-Amiloide/genética , Animales , Humanos , Ratones , Ratones Transgénicos , Microscopía Confocal
14.
Br J Pharmacol ; 165(5): 1442-56, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21883150

RESUMEN

BACKGROUND AND PURPOSE: ß-Adrenoceptor stimulation induces glucose uptake in several insulin-sensitive tissues by poorly understood mechanisms. EXPERIMENTAL APPROACH: We used a model system in CHO-K1 cells expressing the human ß(2)-adrenoceptor and glucose transporter 4 (GLUT4) to investigate the signalling mechanisms involved. KEY RESULTS: In CHO-K1 cells, there was no response to ß-adrenoceptor agonists. The introduction of ß(2)-adrenoceptors and GLUT4 into these cells caused increased glucose uptake in response to ß-adrenoceptor agonists. GLUT4 translocation occurred in response to insulin and ß(2)-adrenoceptor stimulation, although the key insulin signalling intermediate PKB was not phosphorylated in response to ß(2)-adrenoceptor stimulation. Truncation of the C-terminus of the ß(2)-adrenoceptor at position 349 to remove known phosphorylation sites for GPCR kinases (GRKs) or at position 344 to remove an additional PKA site together with the GRK phosphorylation sites did not significantly affect cAMP accumulation but decreased ß(2)-adrenoceptor-stimulated glucose uptake. Furthermore, inhibition of GRK by transfection of the ßARKct construct inhibited ß(2)-adrenoceptor-mediated glucose uptake and GLUT4 translocation, and overexpression of a kinase-dead GRK2 mutant (GRK2 K220R) also inhibited GLUT4 translocation. Introducing ß(2)-adrenoceptors lacking phosphorylation sites for GRK or PKA demonstrated that the GRK sites, but not the PKA sites, were necessary for GLUT4 translocation. CONCLUSIONS AND IMPLICATIONS: Glucose uptake in response to activation of ß(2)-adrenoceptors involves translocation of GLUT4 in this model system. The mechanism is dependent on the C-terminus of the ß(2)-adrenoceptor, requires GRK phosphorylation sites, and involves a signalling pathway distinct from that stimulated by insulin.


Asunto(s)
Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Células CHO , Células Cultivadas , Cricetinae , AMP Cíclico/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Humanos , Insulina/metabolismo , Péptidos/metabolismo , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptores Adrenérgicos beta 2/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos
15.
PLoS One ; 6(7): e22304, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829455

RESUMEN

The ß-adrenoceptors (ß-ARs) control many cellular processes. Here, we show that ß-ARs inhibit calcium depletion-induced cell contractility and subsequent cell detachment of L6 skeletal muscle cells. The mechanism underlying the cell detachment inhibition was studied by using a quantitative cell detachment assay. We demonstrate that cell detachment induced by depletion of extracellular calcium is due to myosin- and ROCK-dependent contractility. The ß-AR inhibition of L6 skeletal muscle cell detachment was shown to be mediated by the ß(2)-AR and increased cAMP but was surprisingly not dependent on the classical downstream effectors PKA or Epac, nor was it dependent on PKG, PI3K or PKC. However, inhibition of potassium channels blocks the ß(2)-AR mediated effects. Furthermore, activation of potassium channels fully mimicked the results of ß(2)-AR activation. In conclusion, we present a novel finding that ß(2)-AR signaling inhibits contractility and thus cell detachment in L6 skeletal muscle cells by a cAMP and potassium channel dependent mechanism.


Asunto(s)
Adhesión Celular , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Células CHO , Calcio/metabolismo , Células Cultivadas , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Eritromicina/análogos & derivados , Eritromicina/metabolismo , Contracción Muscular/efectos de los fármacos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Canales de Potasio/metabolismo , Proteína Quinasa C/metabolismo , Ratas , Transducción de Señal , Quinasas Asociadas a rho/metabolismo
16.
Autophagy ; 7(12): 1528-45, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22108004

RESUMEN

Increasing evidence suggests the toxicity of intracellular amyloid ß-protein (Aß) to neurons, as well as the involvement of oxidative stress in Alzheimer disease (AD). Here we show that normobaric hyperoxia (exposure of cells to 40% oxygen for five days), and consequent activation of macroautophagy and accumulation of Aß within lysosomes, induced apoptosis in differentiated SH-SY5Y neuroblastoma cells. Cells under hyperoxia showed: (1) increased numbers of autophagic vacuoles that contained amyloid precursor protein (APP) as well as Aß monomers and oligomers, (2) increased reactive oxygen species production, and (3) enhanced apoptosis. Oxidant-induced apoptosis positively correlated with cellular Aß production, being the highest in cells that were stably transfected with APP Swedish KM670/671NL double mutation. Inhibition of γ-secretase, prior and/or in parallel to hyperoxia, suggested that the increase of lysosomal Aß resulted mainly from its autophagic uptake, but also from APP processing within autophagic vacuoles. The oxidative stress-mediated effects were prevented by macroautophagy inhibition using 3-methyladenine or ATG5 downregulation. Our results suggest that upregulation of macroautophagy and resulting lysosomal Aß accumulation are essential for oxidant-induced apoptosis in cultured neuroblastoma cells and provide additional support for the interactive role of oxidative stress and the lysosomal system in AD-related neurodegeneration.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Lisosomas/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Oxidantes/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proteína 5 Relacionada con la Autofagia , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mutantes/metabolismo , Oxígeno/farmacología , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transfección , Tretinoina/farmacología , Células Tumorales Cultivadas , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo , Vacuolas/ultraestructura
17.
PLoS One ; 6(7): e22510, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21818330

RESUMEN

BACKGROUND: There is considerable interest in identifying compounds that can improve glucose homeostasis. Skeletal muscle, due to its large mass, is the principal organ for glucose disposal in the body and we have investigated here if shikonin, a naphthoquinone derived from the Chinese plant Lithospermum erythrorhizon, increases glucose uptake in skeletal muscle cells. METHODOLOGY/PRINCIPAL FINDINGS: Shikonin increases glucose uptake in L6 skeletal muscle myotubes, but does not phosphorylate Akt, indicating that in skeletal muscle cells its effect is medaited via a pathway distinct from that used for insulin-stimulated uptake. Furthermore we find no evidence for the involvement of AMP-activated protein kinase in shikonin induced glucose uptake. Shikonin increases the intracellular levels of calcium in these cells and this increase is necessary for shikonin-mediated glucose uptake. Furthermore, we found that shikonin stimulated the translocation of GLUT4 from intracellular vesicles to the cell surface in L6 myoblasts. The beneficial effect of shikonin on glucose uptake was investigated in vivo by measuring plasma glucose levels and insulin sensitivity in spontaneously diabetic Goto-Kakizaki rats. Treatment with shikonin (10 mg/kg intraperitoneally) once daily for 4 days significantly decreased plasma glucose levels. In an insulin sensitivity test (s.c. injection of 0.5 U/kg insulin), plasma glucose levels were significantly lower in the shikonin-treated rats. In conclusion, shikonin increases glucose uptake in muscle cells via an insulin-independent pathway dependent on calcium. CONCLUSIONS/SIGNIFICANCE: Shikonin increases glucose uptake in skeletal muscle cells via an insulin-independent pathway dependent on calcium. The beneficial effects of shikonin on glucose metabolism, both in vitro and in vivo, show that the compound possesses properties that make it of considerable interest for developing novel treatment of type 2 diabetes.


Asunto(s)
Glucemia/efectos de los fármacos , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Músculo Esquelético/patología , Naftoquinonas/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/farmacología , Línea Celular , Diabetes Mellitus Experimental/patología , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/farmacología , Masculino , Células Musculares/patología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/enzimología , Naftoquinonas/administración & dosificación , Consumo de Oxígeno/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
18.
Free Radic Biol Med ; 46(3): 422-9, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19038331

RESUMEN

There is increasing evidence for the toxicity of intracellular amyloid beta-protein (Abeta) to neurons and the involvement of lysosomes in this process in Alzheimer disease (AD). We have recently shown that oxidative stress, a recognized determinant of AD, enhances macroautophagy and leads to intralysosomal accumulation of Abeta in cultured neuroblastoma cells. We hypothesized that oxidative stress promotes AD by stimulating macroautophagy of Abeta that further may induce cell death by destabilizing lysosomal membranes. To investigate such possibility, we compared the effects of hyperoxia (40% ambient oxygen) in cultured HEK293 cells that were transfected with an empty vector (Vector), wild-type APP (APPwt), or Swedish mutant APP (APPswe). Exposure to hyperoxia for 5 days increased the number of cells with Abeta-containing lysosomes, as well as the number of apoptotic cells, compared to normoxic conditions. The rate of apoptosis in all three cell lines demonstrated dependence on intralysosomal Abeta content (Vector

Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Lisosomas/metabolismo , Proteínas Mutantes/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Apoptosis/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/genética , Humanos , Membranas Intracelulares/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/genética , Proteínas Mutantes/genética , Neuronas/patología , Estrés Oxidativo , Oxígeno/farmacología , Transfección , Transgenes
20.
FEBS J ; 276(18): 5041-52, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19663908

RESUMEN

Mutations in the E3 ubiquitin ligase parkin cause early-onset, autosomal-recessive juvenile parkinsonism (AJRP), presumably as a result of a lack of function that alters the level, activity, aggregation or localization of its substrates. Recently, we have reported that phospholipase Cgamma1 is a substrate for parkin. In this article, we show that parkin mutants and siRNA parkin knockdown cells possess enhanced levels of phospholipase Cgamma1 phosphorylation, basal phosphoinositide hydrolysis and intracellular Ca2+ concentration. The protein levels of Ca2+-regulated protein kinase Calpha were decreased in AJRP parkin mutant cells. Neomycin and dantrolene both decreased the intracellular Ca2+ levels in parkin mutants in comparison with those seen in wild-type parkin cells, suggesting that the differences were a consequence of altered phospholipase C activity. The protection of wild-type parkin against 6-hydroxydopamine (6OHDA) toxicity was also established in ARJP mutants on pretreatment with dantrolene, implying that a balancing Ca2+ release from ryanodine-sensitive stores decreases the toxic effects of 6OHDA. Our findings suggest that parkin is an important factor for maintaining Ca2+ homeostasis and that parkin deficiency leads to a phospholipase C-dependent increase in intracellular Ca2+ levels, which make cells more vulnerable to neurotoxins, such as 6OHDA.


Asunto(s)
Calcio/metabolismo , Homeostasis , Fosfolipasa C gamma/fisiología , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Línea Celular Tumoral , Dantroleno/farmacología , Humanos , Oxidopamina/toxicidad , Fosfatidilinositoles/metabolismo , Proteína Quinasa C-alfa/fisiología , Ubiquitina-Proteína Ligasas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA