Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Expert Opin Investig Drugs ; 33(3): 183-190, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38372052

ABSTRACT

INTRODUCTION: Type 2 diabetes (T2D) is metabolic disorder associated with a decrease in insulin activity and/or secretion from the ß-cells of the pancreas, leading to elevated circulating glucose. Current management practices for T2D are complex with varying long-term effectiveness. Agonism of the G protein-coupled receptor GPR119 has received a lot of recent interest as a potential T2D therapeutic. AREAS COVERED: This article reviews studies focused on GPR119 agonism in animal models of T2D and in patients with T2D. EXPERT OPINION: GPR119 agonists in vitro and in vivo can potentially regulate incretin hormone release from the gut, then pancreatic insulin release which regulates blood glucose concentrations. However, the success in controlling glucose homeostasis in rodent models of T2D and obesity, failed to translate to early-stage clinical trials in patients with T2D. However, in more recent studies, acute and chronic dosing with the GPR119 agonist DS-8500a had increased efficacy, although this compound was discontinued for further development. New trials on GPR119 agonists are needed, however it may be that the future of GPR119 agonists lie in the development of combination therapy with other T2D therapeutics.


Subject(s)
Diabetes Mellitus, Type 2 , Animals , Humans , Diabetes Mellitus, Type 2/drug therapy , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Incretins , Insulin/metabolism , Receptors, G-Protein-Coupled/agonists
2.
Pharmacol Res Perspect ; 12(1): e1176, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38332691

ABSTRACT

Truncation of the C-terminal tail of the ß2 -AR, transfection of ßARKct or over-expression of a kinase-dead GRK mutant reduces isoprenaline-stimulated glucose uptake, indicating that GRK is important for this response. We explored whether phosphorylation of the ß2 -AR by GRK2 has a role in glucose uptake or if this response is related to the role of GRK2 as a scaffolding protein. CHO-GLUT4myc cells expressing wild-type and mutant ß2 -ARs were generated and receptor affinity for [3 H]-CGP12177A and density of binding sites determined together with the affinity of isoprenaline and BRL37344. Following receptor activation by ß2 -AR agonists, cAMP accumulation, GLUT4 translocation, [3 H]-2-deoxyglucose uptake, and ß2 -AR internalization were measured. Bioluminescence resonance energy transfer was used to investigate interactions between ß2 -AR and ß-arrestin2 or between ß2 -AR and GRK2. Glucose uptake after siRNA knockdown or GRK inhibitors was measured in response to ß2 -AR agonists. BRL37344 was a poor partial agonist for cAMP generation but displayed similar potency and efficacy to isoprenaline for glucose uptake and GLUT4 translocation. These responses to ß2 -AR agonists occurred in CHO-GLUT4myc cells expressing ß2 -ARs lacking GRK or GRK/PKA phosphorylation sites as well as in cells expressing the wild-type ß2 -AR. However, ß2 -ARs lacking phosphorylation sites failed to recruit ß-arrestin2 and did not internalize. GRK2 knock-down or GRK2 inhibitors decreased isoprenaline-stimulated glucose uptake in rat L6 skeletal muscle cells. Thus, GRK phosphorylation of the ß2 -AR is not associated with isoprenaline- or BRL37344-stimulated glucose uptake. However, GRKs acting as scaffold proteins are important for glucose uptake as GRK2 knock-down or GRK2 inhibition reduces isoprenaline-stimulated glucose uptake.


Subject(s)
G-Protein-Coupled Receptor Kinases , Glucose , Rats , Animals , Isoproterenol/pharmacology , Glucose/metabolism , Receptors, G-Protein-Coupled , Receptors, Adrenergic
4.
Int J Mol Sci ; 24(8)2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37108770

ABSTRACT

Diet-induced obesity (DIO) is a contributor to co-morbidities, resulting in alterations in hormones, lipids, and low-grade inflammation, with the cannabinoid type 2 receptor (CB2) contributing to the inflammatory response. The effects of modulating CB2 with pharmacological treatments on inflammation and adaptations to the obese state are not known. Therefore, we aimed to investigate the molecular mechanisms in adipose tissue of CB2 agonism and CB2 antagonism treatment in a DIO model. Male Sprague Dawley rats were placed on a high-fat diet (HFD) (21% fat) for 9 weeks, then received daily intraperitoneal injections with a vehicle, AM630 (0.3 mg/kg), or AM1241 (3 mg/kg), for a further 6 weeks. AM630 or AM1241 treatment in DIO rats did not alter their body weight, food intake, or liver weight, and it had no effect on their numerous circulating cytokines or peri-renal fat pad mass. AM1241 decreased heart weight and BAT weight; both treatments (AM630 or AM1241) decreased plasma leptin levels, while AM630 also decreased plasma ghrelin and GLP-1 levels. Both treatments decreased Adrb3 and TNF-α mRNA levels in eWAT and TNF-α levels in pWAT. AM630 treatment also decreased the mRNA levels of Cnr2, leptin, and Slc2a4 in eWAT. In BAT, both treatments decreased leptin, UCP1, and Slc2a4 mRNA levels, with AM1241 also decreasing Adrb3, IL1ß, and PRDM16 mRNA levels, and AM630 increasing IL6 mRNA levels. In DIO, CB2 agonist and CB2 antagonist treatment reduces circulating leptin in the absence of weight loss and modulates the mRNA responsible for thermogenesis.


Subject(s)
Cannabinoids , Leptin , Rats , Male , Animals , Tumor Necrosis Factor-alpha/adverse effects , RNA, Messenger/genetics , Rats, Sprague-Dawley , Obesity/drug therapy , Obesity/etiology , Adipose Tissue , Cannabinoids/pharmacology , Receptors, Cannabinoid , Diet, High-Fat/adverse effects , Inflammation/chemically induced , Thermogenesis , Receptor, Cannabinoid, CB2/genetics
5.
Br J Pharmacol ; 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37095602

ABSTRACT

Heart failure remains a leading cause of morbidity and mortality worldwide. Current treatment for patients with heart failure include drugs targeting G protein-coupled receptors such as ß-adrenoceptor antagonists (ß-blockers) and angiotensin II type 1 receptor antagonists (or angiotensin II receptor blockers). However, many patients progress to advanced heart failure with persistent symptoms, despite treatment with available therapeutics that have been shown to reduce mortality and mortality. GPCR targets currently being explored for the development of novel heart failure therapeutics include adenosine receptor, formyl peptide receptor, relaxin/insulin-like family peptide receptor, vasopressin receptor, endothelin receptor and the glucagon-like peptide 1 receptor. Many GPCR drug candidates are limited by insufficient efficacy and/or dose-limiting unwanted effects. Understanding the current challenges hindering successful clinical translation and the potential to overcome existing limitations will facilitate the future development of novel heart failure therapeutics.

6.
Int J Mol Sci ; 23(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36232744

ABSTRACT

Diet-induced obesity (DIO) reduces fatty acid oxidation in skeletal muscle and decreases circulating levels of adiponectin. Endocannabinoid signaling is overactive in obesity, with some effects abated by antagonism of cannabinoid receptor 1 (CB1). This research aimed to determine if treatment with the global CB1 antagonist/inverse agonist, AM251, in high-fat diet (HFD) fed rats influenced adiponectin signaling in skeletal muscle and a "browning" of white adipose tissue (WAT) defined by UCP1 expression levels. Male Sprague Dawley rats consumed an HFD (21% fat) for 9 weeks before receiving daily intraperitoneal injections with vehicle or AM251 (3 mg/kg) for 6 weeks. mRNA expression of genes involved in metabolic functions were measured in skeletal muscle and adipose tissue, and blood was harvested for the measurement of hormones and cytokines. Muscle citrate synthase activity was also measured. AM251 treatment decreased fat pad weight (epididymal, peri-renal, brown), and plasma levels of leptin, glucagon, ghrelin, and GLP-1, and increased PAI-1 along with a range of pro-inflammatory and anti-inflammatory cytokines; however, AM251 did not alter plasma adiponectin levels, skeletal muscle citrate synthase activity or mRNA expression of the genes measured in muscle. AM251 treatment had no effect on white fat UCP1 expression levels. AM251 decreased fat pad mass, altered plasma hormone levels, but did not induce browning of WAT defined by UCP1 mRNA levels or alter gene expression in muscle treated acutely with adiponectin, demonstrating the complexity of the endocannabinoid system and metabolism. The CB1 ligand AM251 increased systemic inflammation suggesting limitations on its use in metabolic disorders.


Subject(s)
Ghrelin , Leptin , Adiponectin/metabolism , Adipose Tissue/metabolism , Animals , Citrate (si)-Synthase/metabolism , Cytokines/metabolism , Diet, High-Fat/adverse effects , Endocannabinoids/metabolism , Ghrelin/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Inflammation/metabolism , Leptin/metabolism , Ligands , Male , Obesity/etiology , Obesity/metabolism , Piperidines , Plasminogen Activator Inhibitor 1/metabolism , Pyrazoles , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptors, Cannabinoid/metabolism , Weight Loss
7.
Biochem Pharmacol ; 188: 114560, 2021 06.
Article in English | MEDLINE | ID: mdl-33844984

ABSTRACT

Emerging evidence suggests that G protein coupled receptor 55 (GPR55) may influence adrenoceptor function/activity in the cardiovascular system. Whether this reflects direct interaction (dimerization) between receptors or signalling crosstalk has not been investigated. This study explored the interaction between GPR55 and the alpha 1A-adrenoceptor (α1A-AR) in the cardiovascular system and the potential to influence function/signalling activities. GPR55 and α1A-AR mediated changes in both cardiac and vascular function was assessed in male wild-type (WT) and GPR55 homozygous knockout (GPR55-/-) mice by pressure volume loop analysis and isolated vessel myography, respectively. Dimerization of GPR55 with the α1A-AR was examined in transfected Chinese hamster ovary-K1 (CHO-K1) cells via Bioluminescence Resonance Energy Transfer (BRET). GPR55 and α1A-AR mediated signalling (extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation) was investigated in neonatal rat ventricular cardiomyocytes using AlphaScreen proximity assays. GPR55-/- mice exhibited both enhanced pressor and inotropic responses to A61603 (α1A-AR agonist), while in isolated vessels, A61603 induced vasoconstriction was attenuated by a GPR55-dependent mechanism. Conversely, GPR55-mediated vasorelaxation was not altered by pharmacological blockade of α1A-ARs with tamsulosin. While cellular studies demonstrated that GPR55 and α1A-AR failed to dimerize, pharmacological blockade of GPR55 altered α1A-AR mediated signalling and reduced ERK1/2 phosphorylation. Taken together, this study provides evidence that GPR55 and α1A-AR do not dimerize to form heteromers, but do interact at the signalling level to modulate the function of α1A-AR in the cardiovascular system.


Subject(s)
Protein Multimerization/physiology , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Cannabinoid/deficiency , Receptors, Cannabinoid/genetics , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Animals, Newborn , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Pregnancy , Protein Multimerization/drug effects , Rats , Rats, Sprague-Dawley
8.
Br J Pharmacol ; 178(8): 1789-1804, 2021 04.
Article in English | MEDLINE | ID: mdl-33506492

ABSTRACT

BACKGROUND AND PURPOSE: The physiological role of vascular ß3 -adrenoceptors is not fully understood. Recent evidence suggests cardiac ß3 -adrenoceptors are functionally effective after down-regulation of ß1 /ß2 -adrenoceptors. The functional interaction between the ß3 -adrenoceptor and other ß-adrenoceptor subtypes in rat striated muscle arteries was investigated. EXPERIMENTAL APPROACH: Studies were performed in cremaster muscle arteries isolated from male Sprague-Dawley rats. ß-adrenoceptor expression was assessed through RT-PCR and immunofluorescence. Functional effects of ß3 -adrenoceptor agonists and antagonists and other ß-adrenoceptor ligands were measured using pressure myography. KEY RESULTS: All three ß-adrenoceptor subtypes were present in the endothelium of the cremaster muscle artery. The ß3 -adrenoceptor agonists mirabegron and CL 316,243 had no effect on the diameter of pressurized (70 mmHg) cremaster muscle arterioles with myogenic tone, while the ß3 -adrenoceptor agonist SR 58611A and the nonselective ß-adrenoceptor agonist isoprenaline caused concentration-dependent dilation. In the presence of ß1/2 -adrenoceptor antagonists nadolol (10 µM), atenolol (1 µM) and ICI 118,551 (0.1 µM) both mirabegron and CL 316,243 were effective in causing vasodilation and the potency of SR 58611A was enhanced, while responses to isoprenaline were inhibited. The ß3 -adrenoceptor antagonist L 748,337 (1 µM) inhibited vasodilation caused by ß3 -adrenoceptor agonists (in the presence of ß1/2 -adrenoceptor blockade), but L 748,337 had no effect on isoprenaline-induced vasodilation. CONCLUSION AND IMPLICATIONS: All three ß-adrenoceptor subtypes were present in the endothelium of the rat cremaster muscle artery, but ß3 -adrenoceptor mediated vasodilation was only evident after blockade of ß1/2 -adrenoceptors. This suggests constitutive ß1/2 -adrenoceptor activity inhibits ß3 -adrenoceptor function in the endothelium of skeletal muscle resistance arteries.


Subject(s)
Abdominal Muscles/blood supply , Adrenergic beta-Antagonists , Arteries/physiology , Receptors, Adrenergic, beta-2 , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Arterioles , Male , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta , Receptors, Adrenergic, beta-3
9.
Pharmacol Res Perspect ; 8(5): e00643, 2020 10.
Article in English | MEDLINE | ID: mdl-32813332

ABSTRACT

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.


Subject(s)
Acetanilides/administration & dosage , Adipocytes, Beige/metabolism , Adipocytes, Brown/metabolism , Adrenergic beta-3 Receptor Agonists/administration & dosage , Glycolysis/drug effects , Thiazoles/administration & dosage , Uncoupling Protein 1/genetics , Acetanilides/pharmacology , Adenosine Monophosphate/metabolism , Adipocytes, Beige/drug effects , Adipocytes, Brown/drug effects , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , CHO Cells , Cells, Cultured , Cricetulus , Deoxyglucose/metabolism , Gene Knockout Techniques , Male , Mice , Oxygen/metabolism , Thiazoles/pharmacology
10.
Mol Metab ; 30: 240-249, 2019 12.
Article in English | MEDLINE | ID: mdl-31767175

ABSTRACT

OBJECTIVE: ß-adrenoceptor mediated activation of brown adipose tissue (BAT) has been associated with improvements in metabolic health in models of type 2 diabetes and obesity due to its unique ability to increase whole body energy expenditure, and rate of glucose and free fatty acid disposal. While the thermogenic arm of this phenomenon has been studied in great detail, the underlying mechanisms involved in ß-adrenoceptor mediated glucose uptake in BAT are relatively understudied. As ß-adrenoceptor agonist administration results in increased hepatic gluconeogenesis that can consequently result in secondary pancreatic insulin release, there is uncertainty regarding the importance of insulin and the subsequent activation of its downstream effectors in mediating ß-adrenoceptor stimulated glucose uptake in BAT. Therefore, in this study, we made an effort to discriminate between the two pathways and address whether the insulin signaling pathway is dispensable for the effects of ß-adrenoceptor activation on glucose uptake in BAT. METHODS: Using a specific inhibitor of phosphoinositide 3-kinase α (PI3Kα), which effectively inhibits the insulin signaling pathway, we examined the effects of various ß-adrenoceptor agonists, including the physiological endogenous agonist norepinephrine on glucose uptake and respiration in mouse brown adipocytes in vitro and on glucose clearance in mice in vivo. RESULTS: PI3Kα inhibition in mouse primary brown adipocytes in vitro, did not inhibit ß-adrenoceptor stimulated glucose uptake, GLUT1 synthesis, GLUT1 translocation or respiration. Furthermore, ß-adrenoceptor mediated glucose clearance in vivo did not require insulin or Akt activation but was attenuated upon administration of a ß3-adrenoceptor antagonist. CONCLUSIONS: We conclude that the ß-adrenergic pathway is still functionally intact upon the inhibition of PI3Kα, showing that the activation of downstream insulin effectors is not required for the acute effects of ß-adrenoceptor agonists on glucose homeostasis or thermogenesis.


Subject(s)
Adipose Tissue, Brown/metabolism , Glucose/metabolism , Receptors, Adrenergic, beta/metabolism , Adipocytes, Brown/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism , Epinephrine/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Norepinephrine/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/metabolism , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , Thermogenesis , Uncoupling Protein 1/metabolism
11.
Sci Signal ; 12(596)2019 08 27.
Article in English | MEDLINE | ID: mdl-31455725

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that play a major role in immunosurveillance against tumor initiation and metastatic spread. The signals and checkpoints that regulate NK cell fitness and function in the tumor microenvironment are not well defined. Transforming growth factor-ß (TGF-ß) is a suppressor of NK cells that inhibits interleukin-15 (IL-15)-dependent signaling events and increases the abundance of receptors that promote tissue residency. Here, we showed that NK cells express the type I activin receptor ALK4, which, upon binding to its ligand activin-A, phosphorylated SMAD2/3 to suppress IL-15-mediated NK cell metabolism. Activin-A impaired human and mouse NK cell proliferation and reduced the production of granzyme B to impair tumor killing. Similar to TGF-ß, activin-A also induced SMAD2/3 phosphorylation and stimulated NK cells to increase their cell surface expression of several markers of ILC1 cells. Activin-A also induced these changes in TGF-ß receptor-deficient NK cells, suggesting that activin-A and TGF-ß stimulate independent pathways that drive SMAD2/3-mediated NK cell suppression. Last, inhibition of activin-A by follistatin substantially slowed orthotopic melanoma growth in mice. These data highlight the relevance of examining TGF-ß-independent SMAD2/3 signaling mechanisms as a therapeutic axis to relieve NK cell suppression and promote antitumor immunity.


Subject(s)
Activins/antagonists & inhibitors , Follistatin/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Signal Transduction/drug effects , Activins/metabolism , Animals , Killer Cells, Natural , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology
12.
BMC Cancer ; 19(1): 157, 2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30777055

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common and aggressive type of primary brain cancer. With median survival of less than 15 months, identification and validation of new GBM therapeutic targets is of critical importance. RESULTS: In this study we tested expression and performed pharmacological characterization of the calcitonin receptor (CTR) as well as other members of the calcitonin family of receptors in high-grade glioma (HGG) cell lines derived from individual patient tumours, cultured in defined conditions. Previous immunohistochemical data demonstrated CTR expression in GBM biopsies and we were able to confirm CALCR (gene encoding CTR) expression. However, as assessed by cAMP accumulation assay, only one of the studied cell lines expressed functional CTR, while the other cell lines have functional CGRP (CLR/RAMP1) receptors. The only CTR-expressing cell line (SB2b) showed modest coupling to the cAMP pathway and no activation of other known CTR signaling pathways, including ERK1/2 and p38 MAP kinases, and Ca2+ mobilization, supportive of low cell surface receptor expression. Exome sequencing data failed to account for the discrepancy between functional data and expression on the cell lines that do not respond to calcitonin(s) with no deleterious non-synonymous polymorphisms detected, suggesting that other factors may be at play, such as alternative splicing or rapid constitutive receptor internalisation. CONCLUSIONS: This study shows that GPCR signaling can display significant variation depending on cellular system used, and effects seen in model recombinant cell lines or tumour cell lines are not always reproduced in a more physiologically relevant system and vice versa.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Cell Culture Techniques , Cell Proliferation , Glioblastoma/mortality , Humans , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptor Activity-Modifying Protein 1/genetics , Receptor Activity-Modifying Protein 2/genetics , Signal Transduction , Survival Analysis , Transcriptome , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Br J Pharmacol ; 176(14): 2416-2432, 2019 07.
Article in English | MEDLINE | ID: mdl-30801689

ABSTRACT

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.


Subject(s)
Adipocytes, Beige/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Receptors, Adrenergic/metabolism , Animals , Humans
14.
Br J Pharmacol ; 176(14): 2433-2448, 2019 07.
Article in English | MEDLINE | ID: mdl-30740664

ABSTRACT

A vital role of adrenoceptors in metabolism and energy balance has been well documented in the heart, skeletal muscle, and adipose tissue. It has been only recently demonstrated, however, that activation of the mechanistic target of rapamycin (mTOR) makes a significant contribution to various metabolic and physiological responses to adrenoceptor agonists. mTOR exists as two distinct complexes named mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) and has been shown to play a critical role in protein synthesis, cell proliferation, hypertrophy, mitochondrial function, and glucose uptake. This review will describe the physiological significance of mTORC1 and 2 as a novel paradigm of adrenoceptor signalling in the heart, skeletal muscle, and adipose tissue. Understanding the detailed signalling cascades of adrenoceptors and how they regulate physiological responses is important for identifying new therapeutic targets and identifying novel therapeutic interventions. 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.


Subject(s)
Adipose Tissue/metabolism , Muscle, Skeletal/metabolism , Sirolimus/metabolism , Adipose Tissue/drug effects , Adrenergic beta-Agonists/pharmacology , Animals , Humans , Muscle, Skeletal/drug effects , Sirolimus/agonists
15.
J Appl Physiol (1985) ; 125(5): 1368-1377, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30138082

ABSTRACT

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.


Subject(s)
Hypoxia/metabolism , Infant, Premature/metabolism , Ischemia/metabolism , Myocardium/metabolism , Receptors, Adrenergic/metabolism , Animals , Animals, Newborn , Cardiotonic Agents , Dobutamine , Dopamine , Heart/drug effects , Models, Animal , Sheep
16.
Article in English | MEDLINE | ID: mdl-29910772

ABSTRACT

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.

17.
J Mol Endocrinol ; 60(3): 213-224, 2018 04.
Article in English | MEDLINE | ID: mdl-29535183

ABSTRACT

Insulin-like peptide 5 (INSL5) is a newly discovered gut hormone expressed in colonic enteroendocrine L-cells but little is known about its biological function. Here, we show using RT-qPCR and in situ hybridisation that Insl5 mRNA is highly expressed in the mouse colonic mucosa, colocalised with proglucagon immunoreactivity. In comparison, mRNA for RXFP4 (the cognate receptor for INSL5) is expressed in various mouse tissues, including the intestinal tract. We show that the human enteroendocrine L-cell model NCI-H716 cell line, and goblet-like colorectal cell lines SW1463 and LS513 endogenously express RXFP4. Stimulation of NCI-H716 cells with INSL5 produced phosphorylation of ERK1/2 (Thr202/Tyr204), AKT (Thr308 and Ser473) and S6RP (Ser235/236) and inhibited cAMP production but did not stimulate Ca2+ release. Acute INSL5 treatment had no effect on GLP-1 secretion mediated by carbachol or insulin, but modestly inhibited forskolin-stimulated GLP-1 secretion in NCI-H716 cells. However, chronic INSL5 pre-treatment (18 h) increased basal GLP-1 secretion and prevented the inhibitory effect of acute INSL5 administration. LS513 cells were found to be unresponsive to INSL5 despite expressing RXFP4 Another enteroendocrine L-cell model, mouse GLUTag cells did not express detectable levels of Rxfp4 and were unresponsive to INSL5. This study provides novel insights into possible autocrine/paracrine roles of INSL5 in the intestinal tract.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Insulin/metabolism , Proteins/metabolism , Signal Transduction , Animals , Cell Line , Colon/metabolism , Cyclic AMP/biosynthesis , Gene Expression Profiling , Goblet Cells/metabolism , Humans , Insulin/genetics , Mice, Inbred C57BL , Phosphorylation , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism
18.
Drug Discov Today ; 23(3): 592-604, 2018 03.
Article in English | MEDLINE | ID: mdl-29331500

ABSTRACT

Dysfunction of the endocannabinoid system (ECS) has been identified in metabolic disease. Cannabinoid receptor 1 (CB1) is abundantly expressed in the brain but also expressed in the periphery. Cannabinoid receptor 2 (CB2) is more abundant in the periphery, including the immune cells. In obesity, global antagonism of overexpressed CB1 reduces bodyweight but leads to centrally mediated adverse psychological outcomes. Emerging research in isolated cultured cells or tissues has demonstrated that targeting the endocannabinoid system in the periphery alleviates the pathologies associated with metabolic disease. Further, peripheral specific cannabinoid ligands can reverse aspects of the metabolic phenotype. This Keynote review will focus on current research on the functionality of peripheral modulation of the ECS for the treatment of obesity.


Subject(s)
Endocannabinoids/metabolism , Metabolic Diseases/metabolism , Animals , Humans , Obesity/metabolism , Receptors, Cannabinoid/metabolism
19.
Biochem Pharmacol ; 148: 27-40, 2018 02.
Article in English | MEDLINE | ID: mdl-29175420

ABSTRACT

The capacity of G protein-coupled receptors to modulate mechanistic target of rapamycin (mTOR) activity is a newly emerging paradigm with the potential to link cell surface receptors with cell survival. Cardiomyocyte viability is linked to signalling pathways involving Akt and mTOR, as well as increased glucose uptake and utilization. Our aim was to determine whether the α1A-adrenoceptor (AR) couples to these protective pathways, and increased glucose uptake. We characterised α1A-AR signalling in CHO-K1 cells co-expressing the human α1A-AR and GLUT4 (CHOα1AGLUT4myc) and in neonatal rat ventricular cardiomyocytes (NRVM), and measured glucose uptake, intracellular Ca2+ mobilization, and phosphorylation of mTOR, Akt, 5' adenosine monophosphate-activated kinase (AMPK) and S6 ribosomal protein (S6rp). In both systems, noradrenaline and the α1A-AR selective agonist A61603 stimulated glucose uptake by parallel pathways involving mTOR and AMPK, whereas another α1-AR agonist oxymetazoline increased glucose uptake predominantly by mTOR. All agonists promoted phosphorylation of mTOR at Ser2448 and Ser2481, indicating activation of both mTORC1 and mTORC2, but did not increase Akt phosphorylation. In CHOα1AGLUT4myc cells, siRNA directed against rictor but not raptor suppressed α1A-AR mediated glucose uptake. We have thus identified mTORC2 as a key component in glucose uptake stimulated by α1A-AR agonists. Our findings identify a novel link between the α1A-AR, mTORC2 and glucose uptake, that have been implicated separately in cardiomyocyte survival. Our studies provide an improved framework for examining the utility of α1A-AR selective agonists as tools in the treatment of cardiac dysfunction.


Subject(s)
Glucose/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, alpha-1/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , CHO Cells , Calcimycin , Calcium , Cricetinae , Cricetulus , Gene Expression Regulation/drug effects , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Norepinephrine , Phosphorylation , Prazosin/metabolism , Prazosin/pharmacology , Rats , Signal Transduction , TOR Serine-Threonine Kinases/genetics
20.
Cell Signal ; 42: 54-66, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28970184

ABSTRACT

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.


Subject(s)
Adipocytes, Beige/drug effects , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Mitochondria/drug effects , Receptors, Adrenergic, beta-3/genetics , Thiazolidinediones/pharmacology , Adipocytes, Beige/cytology , Adipocytes, Beige/metabolism , Adipocytes, Brown/cytology , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipocytes, White/cytology , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Animals , Biological Transport , Cyclic AMP/agonists , Cyclic AMP/metabolism , Gene Expression Regulation , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Male , Mice , Mitochondria/metabolism , Organ Specificity , Oxygen Consumption/drug effects , PPAR gamma/agonists , PPAR gamma/genetics , PPAR gamma/metabolism , Primary Cell Culture , Receptors, Adrenergic, beta-3/metabolism , Rosiglitazone , Signal Transduction , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...