Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Pharmacol ; 101(4): 246-256, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35125345

RESUMEN

Specialized proresolving mediators (SPMs) and their cognate G protein-coupled receptors are implicated in autoimmune disorders, including chronic inflammation, rheumatoid arthritis, systemic scleroderma, and lupus erythematosus. To date, six G protein-coupled receptors (GPCRs) have been paired with numerous endogenous and synthetic ligands. However, the function and downstream signaling of these receptors remains unclear. To address this knowledge gap, we systematically expressed each receptor in a human embryonic kindney 293 (HEK293)-Flp-In-CD8a-FLAG cell system. Each receptor was pharmacologically characterized with both synthetic and putative endogenous ligands across different signaling assays, covering both G protein-dependent (Gs, Gi, and Gq) and independent mechanisms (ß-arrestin2 recruitment). Three orphan GPCRs previously identified as SPM receptors (GPR 18, GPR32 and GPR37) failed to express in HEK 293 cells. Although we were unsuccessful in identifying an endogenous ligand for formyl peptide receptor 2 (FPR2)/lipoxin A4 receptor (ALX), with only a modest response to N-formylmethionine-leucyl-phenylalanine (fMLP), we did reveal clear signaling bias away from extracelluar signal-related kinase (ERK) 1/2 phosphorylation for the clinically tested agonist N-(2-{[4-(1,1-difluoroethyl)-1,3-oxazol-2-yl]methyl}-2H-1,2,3-triazol-4-yl)-2-methyl-5-(3-methylphenyl)-1,3-oxazole-4-carboxamide (ACT-389949), adding further evidence for its poor efficacy in two phase I studies. We also identified neuroprotectin D1 as a new leukotriene B4 receptor 1 (BLT1) agonist, implying an alternative target for the neuroprotective effects of the ligand. We confirmed activity for resolvin E1 (RvE1) at BLT1 but failed to observe any response at the chemerin1 receptor. This study provides some much-needed clarity around published receptor-ligand pairings but indicates that the expression and function of these SPM GPCRs remains very much context-dependent. In addition, the identification of signaling bias at FPR2/ALX may assist in guiding design of new FPR2/ALX agonists for the treatment of autoimmune disorders. SIGNIFICANCE STATEMENT: To our knowledge, this is the first study to comprehensibly show how several natural mediators and synthetic ligands signal through three specialized proresolving mediator GPCRs using multiple ligands from different classes across four-six endpoint signaling assays. This study discovers new ligand pairings, refutes others, reveals poly-pharmacology, and identifies biased agonism in formyl peptide receptor 2/lipoxin A4 receptor pharmacology. This study highlights the potential of these receptors in treating specific autoimmune diseases, including rheumatoid arthritis, systemic scleroderma, and systemic lupus erythematosus.


Asunto(s)
Artritis Reumatoide , Enfermedades Autoinmunes , Esclerodermia Sistémica , Células HEK293 , Humanos , Ligandos , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/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.
Biochem Pharmacol ; 199: 114985, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35300966

RESUMEN

G protein-coupled receptors (GPCRs) are the largest class of membrane proteins and in recent years there has been a growing appreciation of the importance in understanding temporal aspects of GPCR behaviour, including the kinetics of ligand binding and downstream receptor mediated signalling. Class B1 GPCRs are activated by peptide agonists and are validated therapeutic targets for numerous diseases. However, the kinetics of ligand binding and how this is linked to downstream activation of signalling cascades is not routinely assessed in development of peptide agonists for this receptor class. The glucagon-like peptide-1 receptor (GLP-1R) is a prototypical class B1 GPCR and a validated target for treatment of global health burdens, including type 2 diabetes and obesity. In this study we examined the kinetics of different steps in GLP-1R activation and subsequent cAMP production mediated by a series of GLP-1R peptide agonists, including the ligand-receptor interaction, ligand-receptor-mediated G protein engagement and conformational change and cAMP production. Our results revealed GLP-1R peptide agonist dissociation kinetics (Koff), but not association kinetics (Kon), were positively correlated with the onset of receptor-G protein coupling/conformational change, onset of cAMP production and duration of cAMP signalling. Thus, this study advances the understanding of molecular events that couple GLP-1R ligand binding to intracellular signaling, with the findings likely to have implications for mechanistic understanding of agonist action at other related class B1 GPCRs.


Asunto(s)
Diabetes Mellitus Tipo 2 , Receptor del Péptido 1 Similar al Glucagón , Diabetes Mellitus Tipo 2/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Cinética , Ligandos , Péptidos/química , Receptores Acoplados a Proteínas G/metabolismo
4.
Mol Pharmacol ; 79(2): 298-307, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20978120

RESUMEN

Although G protein-coupled receptors are often categorized in terms of their primary coupling to a given type of Gα protein subunit, it is now well established that many show promiscuous coupling and activate multiple signaling pathways. Furthermore, some agonists selectively activate signaling pathways by promoting interaction between distinct receptor conformational states and particular Gα subunits or alternative signaling proteins. We have tested the capacity of agonists to stimulate Ca(2+) release, cAMP accumulation, and changes in extracellular acidification rate (ECAR) at the human α(1A)-adrenoceptor. Signaling bias factors were determined by novel application of an operational model of agonism and compared with the reference endogenous agonist norepinephrine; values significantly different from 1.0 indicated an agonist that promoted receptor conformations distinct from that favored by norepinephrine. Oxymetazoline was a full agonist for ECAR and a partial agonist for Ca(2+) release (bias factor 8.2) but failed to stimulate cAMP production. Phenylephrine showed substantial bias toward ECAR versus Ca(2+) release or cAMP accumulation (bias factors 21 and 33, respectively) but did not display bias between Ca(2+) and cAMP pathways. Cirazoline and N-[5-(4,5-dihydro-1H-imidazol-2-yl)-2-hydroxy-5,6,7,8-tetrahydronaphthalen-1-yl]methanesulfonamide (A61603) displayed bias toward cAMP relative to Ca(2+) release (bias factors of 7.4 and 8.6). It is noteworthy that epinephrine, a second endogenous adrenoceptor agonist, did not display bias relative to norepinephrine. Our finding that phenylephrine displayed significant signaling bias, despite being highly similar in structure to epinephrine, indicates that subtle differences in agonist-receptor interaction can affect conformational changes in cytoplasmic domains and thereby modulate the repertoire of effector proteins that are activated.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Animales , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Cartilla de ADN , Humanos , Imidazoles/farmacología , Norepinefrina/farmacología , Fenilefrina/farmacología , Tetrahidronaftalenos/farmacología
5.
J Neurochem ; 117(5): 915-26, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21447002

RESUMEN

In the brain, glycogen is primarily stored in astrocytes where it is regulated by several hormones/neurotransmitters, including noradrenaline that controls glycogen breakdown (in the short term) and synthesis. Here, we have examined the adrenoceptor (AR) subtype that mediates the glycogenic effect of noradrenaline in chick primary astrocytes by the measurement of glycogen turnover (total (14) C incorporation of glucose into glycogen) following noradrenergic activation. Noradrenaline and insulin increased glycogen turnover in a concentration-dependent manner. The effect of noradrenaline was mimicked by stimulation of α(2) -ARs (and to a lesser degree by ß(3) -ARs), but not by stimulation of α(1) -, ß(1) -, or ß(2) -ARs, and occurred only in astrocytes and not neurons. In chick astrocytes, studies using RT-PCR and radioligand binding showed that α(2A) - and α(2C) -AR mRNA and protein were present. α(2) -AR- or insulin-mediated glycogen turnover was inhibited by phosphatidylinositol-3 kinase inhibitors, and both insulin and clonidine caused phosphorylation of Akt and glycogen synthase kinase-3 in chick astrocytes. α(2) -AR but not insulin-mediated glycogen turnover was inhibited by pertussis toxin pre-treatment indicating involvement of Gi/o proteins. These results show that the increase in glycogen turnover caused by noradrenaline is because of activation of α(2) -ARs that increase glycogen turnover in astrocytes utilizing a Gi/o-PI3K pathway.


Asunto(s)
Astrocitos/metabolismo , Glucógeno/metabolismo , Norepinefrina/fisiología , Receptores Adrenérgicos alfa 2/fisiología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Astrocitos/efectos de los fármacos , Biotransformación , Células Cultivadas , Embrión de Pollo , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipoglucemiantes/farmacología , Cinética , Proteína Oncogénica v-akt/metabolismo , Fosforilación , Prosencéfalo/metabolismo , Receptores Adrenérgicos alfa 2/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleótidos/farmacología , Yohimbina
6.
Front Pharmacol ; 12: 628060, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33776771

RESUMEN

Adenosine A1 receptors (A1R) are a potential target for cardiac injury treatment due to their cardioprotective/antihypertrophic actions, but drug development has been hampered by on-target side effects such as bradycardia and altered renal hemodynamics. Biased agonism has emerged as an attractive mechanism for A1R-mediated cardioprotection that is haemodynamically safe. Here we investigate the pre-clinical pharmacology, efficacy and side-effect profile of the A1R agonist neladenoson, shown to be safe but ineffective in phase IIb trials for the treatment of heart failure. We compare this agent with the well-characterized, pan-adenosine receptor (AR) agonist NECA, capadenoson, and the A1R biased agonist VCP746, previously shown to be safe and cardioprotective in pre-clinical models of heart failure. We show that like VCP746, neladenoson is biased away from Ca2+ influx relative to NECA and the cAMP pathway at the A1R, a profile predictive of a lack of adenosine-like side effects. Additionally, neladenoson was also biased away from the MAPK pathway at the A1R. In contrast to VCP746, which displays more 'adenosine-like' signaling at the A2BR, neladenoson was a highly selective A1R agonist, with biased, weak agonism at the A2BR. Together these results show that unwanted hemodynamic effects of A1R agonists can be avoided by compounds biased away from Ca2+ influx relative to cAMP, relative to NECA. The failure of neladenoson to reach primary endpoints in clinical trials suggests that A1R-mediated cAMP inhibition may be a poor indicator of effectiveness in chronic heart failure. This study provides additional information that can aid future screening and/or design of improved AR agonists that are safe and efficacious in treating heart failure in patients.

7.
Artículo en Inglés | MEDLINE | ID: mdl-34782333

RESUMEN

INTRODUCTION: A potential role for the orphan G protein-coupled receptor, GPR21, in linking immune cell infiltration into tissues and obesity-induced insulin resistance has been proposed, although limited studies in mice are complicated by non-selective deletion of Gpr21. RESEARCH DESIGN AND METHODS: We hypothesized that a Gpr21-selective knockout mouse model, coupled with type 2 diabetes patient samples, would clarify these issues and enable clear assessment of GPR21 as a potential therapeutic target. RESULTS: High-fat feeding studies in Gpr21-/- mice revealed improved glucose tolerance and modest changes in inflammatory gene expression. Gpr21-/- monocytes and intraperitoneal macrophages had selectively impaired chemotactic responses to monocyte chemoattractant protein (MCP)-1, despite unaltered expression of Ccr2. Further genotypic analysis revealed that chemotactic impairment was due to dysregulated monocyte polarization. Patient samples revealed elevated GPR21 expression in peripheral blood mononuclear cells in type 2 diabetes, which was correlated with both %HbA1c and fasting plasma glucose levels. CONCLUSIONS: Collectively, human and mouse data suggest that GPR21 influences both glucose homeostasis and MCP-1/CCL2-CCR2-driven monocyte migration. However, a Gpr21-/- bone marrow transplantation and high-fat feeding study in mice revealed no effect on glucose homeostasis, suggesting that there is no (or limited) overlap in the mechanism involved for monocyte-driven inflammation and glucose homeostasis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Animales , Quimiocina CCL2/genética , Diabetes Mellitus Tipo 2/genética , Glucosa , Homeostasis , Humanos , Resistencia a la Insulina/genética , Leucocitos Mononucleares , Ratones , Receptores CCR2/genética , Receptores Acoplados a Proteínas G/genética
8.
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
9.
Br J Pharmacol ; 176(14): 2416-2432, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30801689

RESUMEN

Adrenoceptors play an important role in adipose tissue biology and physiology that includes regulating the synthesis and storage of triglycerides (lipogenesis), the breakdown of stored triglycerides (lipolysis), thermogenesis (heat production), glucose metabolism, and the secretion of adipocyte-derived hormones that can control whole-body energy homeostasis. These processes are regulated by the sympathetic nervous system through actions at different adrenoceptor subtypes expressed in adipose tissue depots. In this review, we have highlighted the role of adrenoceptor subtypes in white, brown, and brite adipocytes in both rodents and humans and have included detailed analysis of adrenoceptor expression in human adipose tissue and clonally derived adipocytes. We discuss important considerations when investigating adrenoceptor function in adipose tissue or adipocytes. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.


Asunto(s)
Adipocitos Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Receptores Adrenérgicos/metabolismo , Animales , Humanos
10.
J Appl Physiol (1985) ; 125(5): 1368-1377, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30138082

RESUMEN

Preterm infants frequently suffer cardiovascular compromise, with hypotension and/or low systemic blood flow, leading to tissue hypoxia-ischemia (HI). Many preterm infants respond inadequately to inotropic treatments using adrenergic agonists such as dobutamine (DB) or dopamine (DA). This may be because of altered cardiac adrenoceptor expression because of tissue HI or prolonged exposure to adrenergic agonists. We assessed the effects of severe HI with and without DB/DA treatment on cardiac adrenoceptor expression in preterm fetal sheep. Fetal sheep (93-95 days) exposed to sham surgery or severe HI induced by umbilical cord occlusion received intravenous DB or saline for 74 h (HI + DB, HI, Sham + DB, Sham). The HI groups were also compared with fetal sheep exposed to HI and DA. Fetal hearts were collected to determine ß-adrenoceptor numbers using [125I]-cyanopindolol binding and mRNA expression of ß1-, ß2-, α1A-, α2A-, or α2B-adrenoceptors. The HI group had increased ß-adrenoceptor numbers compared with all other groups in all four heart chambers ( P < 0.05). This increase in ß-adrenoceptor numbers in the HI group was significantly reduced by DB infusion in all four heart chambers, but DA infusion in the HI group only reduced ß-adrenoceptor numbers in the left atria and ventricle. DB alone did not affect ß-adrenoceptor numbers in the sham animals. Changes in ß1-adrenoceptor mRNA levels trended to parallel the binding results. We conclude that HI upregulates preterm fetal cardiac ß-adrenoceptors, but prolonged exposure to adrenergic agonists downregulates adrenoceptors in the preterm heart exposed to HI and may underpin the frequent failure of inotropic therapy in preterm infants. NEW & NOTEWORTHY This is the first study, to our knowledge, on the effects of hypoxia-ischemia and adrenergic agonists on adrenoceptors in the preterm heart. In fetal sheep, we demonstrate that hypoxia-ischemia increases cardiac ß-adrenoceptor numbers. However, exposure to both hypoxia-ischemia and adrenergic agonists (dobutamine or dopamine) reduces the increase in ß-adrenoceptor numbers, which may underpin the inadequate response in human preterm infants to inotropic therapy using adrenergic agonists. Dobutamine alone does not affect the cardiac adrenoceptors in the sham animals.


Asunto(s)
Hipoxia/metabolismo , Recien Nacido Prematuro/metabolismo , Isquemia/metabolismo , Miocardio/metabolismo , Receptores Adrenérgicos/metabolismo , Animales , Animales Recién Nacidos , Cardiotónicos , Dobutamina , Dopamina , Corazón/efectos de los fármacos , Modelos Animales , Ovinos
11.
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
12.
Artículo en Inglés | MEDLINE | ID: mdl-29910772

RESUMEN

The recruitment of brite (or beige) adipocytes has been advocated as a means to combat obesity, due to their ability to phenotypically resemble brown adipocytes (BA). Lineage studies indicate that brite adipocytes are formed by differentiation of precursor cells or by direct conversion of existing white adipocytes, depending on the adipose depot examined. We have systematically compared the gene expression profile and a functional output (oxygen consumption) in mouse adipocytes cultured from two contrasting depots, namely interscapular brown adipose tissue, and inguinal white adipose tissue (iWAT), following treatment with a known browning agent, the peroxisome proliferator-activated receptor (PPARγ) activator rosiglitazone. Prototypical BA readily express uncoupling protein (UCP)1, and upstream regulators including the ß3-adrenoceptor and transcription factors involved in energy homeostasis. Adipocytes from inguinal WAT display maximal UCP1 expression and mitochondrial uncoupling only when treated with a combination of the PPARγ activator rosiglitazone and a ß3-adrenoceptor agonist. In conclusion, brite adipocytes are fully activated only when a browning agent (rosiglitazone) and a thermogenic agent (ß3-adrenoceptor agonist) are added in combination. The presence of rosiglitazone throughout the 7-day culture period partially masks the effects of ß3-adrenoceptor signaling in inguinal white adipocyte cultures, whereas including rosiglitazone only for the first 3 days promotes robust ß3-adrenoceptor expression and provides an improved window for detection of ß3-adrenoceptor responses.

13.
Br J Pharmacol ; 174(14): 2318-2333, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28444738

RESUMEN

BACKGROUND AND PURPOSE: Agonists acting at GPCRs promote biased signalling via Gα or Gßγ subunits, GPCR kinases and ß-arrestins. Since the demonstration of biased agonism has implications for drug discovery, it is essential to consider confounding factors contributing to bias. We have examined bias at human α1A -adrenoceptors stably expressed at low levels in CHO-K1 cells, identifying off-target effects at endogenous receptors that contribute to ERK1/2 phosphorylation in response to the agonist oxymetazoline. EXPERIMENTAL APPROACH: Intracellular Ca2+ mobilization was monitored in a Flexstation® using Fluo 4-AM. The accumulation of cAMP and ERK1/2 phosphorylation were measured using AlphaScreen® proximity assays, and mRNA expression was measured by RT-qPCR. Ligand bias was determined using the operational model of agonism. KEY RESULTS: Noradrenaline, phenylephrine, methoxamine and A61603 increased Ca2+ mobilization, cAMP accumulation and ERK1/2 phosphorylation. However, oxymetazoline showed low efficacy for Ca+2 mobilization, no effect on cAMP generation and high efficacy for ERK1/2 phosphorylation. The apparent functional selectivity of oxymetazoline towards ERK1/2 was related to off-target effects at 5-HT1B receptors endogenously expressed in CHO-K1 cells. Phenylephrine and methoxamine showed genuine bias towards ERK1/2 phosphorylation compared to Ca2+ and cAMP pathways, whereas A61603 displayed bias towards cAMP accumulation compared to ERK1/2 phosphorylation. CONCLUSION AND IMPLICATIONS: We have shown that while adrenergic agonists display bias at human α1A -adrenoceptors, the marked bias of oxymetazoline for ERK1/2 phosphorylation originates from off-target effects. Commonly used cell lines express a repertoire of endogenous GPCRs that may confound studies on biased agonism at recombinant receptors.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Imidazoles/farmacología , Metoxamina/farmacología , Norepinefrina/farmacología , Fenilefrina/farmacología , Receptores Adrenérgicos alfa 1/metabolismo , Tetrahidronaftalenos/farmacología , Agonistas de Receptores Adrenérgicos alfa 1/química , Animales , Células CHO , Células Cultivadas , Cricetulus , Relación Dosis-Respuesta a Droga , Humanos , Imidazoles/química , Metoxamina/química , Norepinefrina/química , Fenilefrina/química , Relación Estructura-Actividad , Tetrahidronaftalenos/química
14.
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
15.
PLoS One ; 8(7): e68841, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935897

RESUMEN

ρ-Da1a is a three-finger fold toxin from green mamba venom that is highly selective for the α1A-adrenoceptor. This toxin has atypical pharmacological properties, including incomplete inhibition of (3)H-prazosin or (125)I-HEAT binding and insurmountable antagonist action. We aimed to clarify its mode of action at the α1A-adrenoceptor. The affinity (pKi 9.26) and selectivity of ρ-Da1a for the α1A-adrenoceptor were confirmed by comparing binding to human adrenoceptors expressed in eukaryotic cells. Equilibrium and kinetic binding experiments were used to demonstrate that ρ-Da1a, prazosin and HEAT compete at the α1A-adrenoceptor. ρ-Da1a did not affect the dissociation kinetics of (3)H-prazosin or (125)I-HEAT, and the IC50 of ρ-Da1a, determined by competition experiments, increased linearly with the concentration of radioligands used, while the residual binding by ρ-Da1a remained stable. The effect of ρ-Da1a on agonist-stimulated Ca(2+) release was insurmountable in the presence of phenethylamine- or imidazoline-type agonists. Ten mutations in the orthosteric binding pocket of the α1A-adrenoceptor were evaluated for alterations in ρ-Da1a affinity. The D106(3.32)A and the S188(5.42)A/S192(5.46)A receptor mutations reduced toxin affinity moderately (6 and 7.6 times, respectively), while the F86(2.64)A, F288(6.51)A and F312(7.39)A mutations diminished it dramatically by 18- to 93-fold. In addition, residue F86(2.64) was identified as a key interaction point for (125)I-HEAT, as the variant F86(2.64)A induced a 23-fold reduction in HEAT affinity. Unlike the M1 muscarinic acetylcholine receptor toxin MT7, ρ-Da1a interacts with the human α1A-adrenoceptor orthosteric pocket and shares receptor interaction points with antagonist (F86(2.64), F288(6.51) and F312(7.39)) and agonist (F288(6.51) and F312(7.39)) ligands. Its selectivity for the α1A-adrenoceptor may result, at least partly, from its interaction with the residue F86(2.64), which appears to be important also for HEAT binding.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/química , Antagonistas de Receptores Adrenérgicos alfa 1/química , Venenos Elapídicos/química , Prazosina/química , Receptores Adrenérgicos alfa 1/química , Tetralonas/química , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Sitios de Unión , Unión Competitiva , Células CHO , Cricetulus , Venenos Elapídicos/farmacología , Elapidae/metabolismo , Humanos , Cinética , Ligandos , Modelos Moleculares , Mutación , Prazosina/farmacología , Unión Proteica , Ensayo de Unión Radioligante , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Tetralonas/farmacología
16.
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
17.
Cell Signal ; 22(7): 1104-13, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20206685

RESUMEN

The role of muscarinic acetylcholine receptors (mAChRs) in regulating glucose uptake in L6 skeletal muscle cells was investigated. [(3)H]-2-Deoxyglucose uptake was increased in differentiated L6 cells by insulin, acetylcholine, oxotremorine-M and carbachol. mAChR-mediated glucose uptake was inhibited by the AMPK inhibitor Compound C. Whole cell radioligand binding using [(3)H]-N-methyl scopolamine chloride identified mAChRs in differentiated but not undifferentiated L6 cells and M(3) mAChR mRNA was detected only in differentiated cells. M(3) mAChRs are Gq-coupled, and cholinergic stimulation by the mAChR agonists acetylcholine, oxotremorine-M and carbachol increased Ca(2+) in differentiated but not undifferentiated L6 cells. This was due to muscarinic but not nicotinic activation as responses were antagonised by the muscarinic antagonist atropine but not the nicotinic antagonist tubocurarine. Western blotting showed that both carbachol and the AMPK activator AICAR increased phosphorylation of the AMPKalpha subunit at Thr172, with responses to carbachol blocked by Compound C and the CaMKK inhibitor STO609 but not by the PI3K inhibitor wortmannin. AICAR-stimulated AMPK phosphorylation was not sensitive to STO-609, confirming that this compound inhibits CaMKK but not the classical AMPK kinase LKB1. The TAK1 inhibitor (5Z)-7-oxozeaenol and the G(i) inhibitor pertussis toxin both failed to block AMPK phosphorylation in response to carbachol. Using CHO-K1 cells stably expressing each of the mAChR subtypes (M(1)-M(4)), it was determined that only the M(1) and M(3) mAChRs phosphorylate AMPK, confirming a G(q)-dependent mechanism. This study demonstrates that activation of M(3) mAChRs in L6 skeletal muscle cells stimulates glucose uptake via a CaMKK-AMPK-dependent mechanism, independent of the insulin-stimulated pathway.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Glucosa/metabolismo , Músculo Esquelético/metabolismo , Proteínas Quinasas/metabolismo , Receptor Muscarínico M3/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico , Células CHO , Calcio/metabolismo , Diferenciación Celular , Cricetinae , Cricetulus , Desoxiglucosa/metabolismo , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA