Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Metab ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871982

ABSTRACT

Incretin-based therapies are highly successful in combatting obesity and type 2 diabetes1. Yet both activation and inhibition of the glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) in combination with glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) activation have resulted in similar clinical outcomes, as demonstrated by the GIPR-GLP-1R co-agonist tirzepatide2 and AMG-133 (ref. 3) combining GIPR antagonism with GLP-1R agonism. This underlines the importance of a better understanding of the GIP system. Here we show the necessity of ß-arrestin recruitment for GIPR function, by combining in vitro pharmacological characterization of 47 GIPR variants with burden testing of clinical phenotypes and in vivo studies. Burden testing of variants with distinct ligand-binding capacity, Gs activation (cyclic adenosine monophosphate production) and ß-arrestin 2 recruitment and internalization shows that unlike variants solely impaired in Gs signalling, variants impaired in both Gs and ß-arrestin 2 recruitment contribute to lower adiposity-related traits. Endosomal Gs-mediated signalling of the variants shows a ß-arrestin dependency and genetic ablation of ß-arrestin 2 impairs cyclic adenosine monophosphate production and decreases GIP efficacy on glucose control in male mice. This study highlights a crucial impact of ß-arrestins in regulating GIPR signalling and overall preservation of biological activity that may facilitate new developments in therapeutic targeting of the GIPR system.

2.
Sci Adv ; 10(10): eadj3823, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38446876

ABSTRACT

Mutations that perturb leptin-melanocortin signaling are known to cause hyperphagia and obesity, but energy expenditure has not been well studied outside rodents. We report on a common canine mutation in pro-opiomelanocortin (POMC), which prevents production of ß-melanocyte-stimulating hormone (ß-MSH) and ß-endorphin but not α-MSH; humans, similar to dogs, produce α-MSH and ß-MSH from the POMC propeptide, but rodents produce only α-MSH. We show that energy expenditure is markedly lower in affected dogs, which also have increased motivational salience in response to a food cue, indicating increased wanting or hunger. There was no difference in satiety at a modified ad libitum meal or in their hedonic response to food, nor disruption of adrenocorticotropic hormone (ACTH) or thyroid axes. In vitro, we show that ß-MSH signals comparably to α-MSH at melanocortin receptors. These data implicate ß-MSH and ß-endorphin as important in determining hunger and moderating energy expenditure and suggest that this role is independent of the presence of α-MSH.


Subject(s)
beta-Endorphin , beta-MSH , Humans , Dogs , Animals , beta-Endorphin/genetics , Basal Metabolism , Pro-Opiomelanocortin/genetics , Hunger , alpha-MSH/genetics
3.
J Endocrinol ; 261(2)2024 May 01.
Article in English | MEDLINE | ID: mdl-38451873

ABSTRACT

The glucagon-like peptide 1 receptor (GLP-1R) is a class B G protein-coupled receptor (GPCR) that emerged as a pharmacologic target in cardiometabolic disease, including diabetes and obesity, over 30 years ago. The subsequent widespread clinical use of GLP-1R agonists, including exenatide, liraglutide, and semaglutide, has made the GLP-1R a preeminent model for understanding basic GPCR biology, including the emergent field of biased agonism. Recent data demonstrate that the dual GLP-1R/glucose dependent insulinotropic polypeptide receptor (GIPR) agonist tirzepatide exhibits a biased signaling profile characterized by preferential Gαs activation over ß-arrestin recruitment, which appears to contribute to its insulinotropic and body-weight reducing effects in preclinical models. This constitutes a major finding in which nuanced, mechanistic receptor signaling dynamics in vitro mediate real-world clinical differentiation within a drug class. Because of the striking bench-top-to-bed side relevance of this biased signaling phenomenon, we have undertaken a review of the emerging data detailing biased agonism at the GLP-1R. In this review, we introduce the core concept of biased agonism followed by a detailed consideration of the key mechanisms, including ligand-mediated bias, receptor-mediated bias, and systems/cell-type bias. Current industry programs are largely, if not entirely, focused on developing biased ligands, and so we have dedicated a section of the review to a brief meta-analysis of compounds reported to drive biased signaling, with a consideration of the structural determinants of receptor-ligand interactions. In this work, we aim to assess the current knowledge regarding signaling bias at the GLP-1R and how these ideas might be leveraged in future optimization.


Subject(s)
Liraglutide , Receptors, G-Protein-Coupled , Ligands , Liraglutide/pharmacology , Exenatide/pharmacology , Signal Transduction , Glucagon-Like Peptide-1 Receptor/agonists
4.
Nat Commun ; 14(1): 1450, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36922513

ABSTRACT

Disruption of brain-expressed G protein-coupled receptor-10 (GPR10) causes obesity in animals. Here, we identify multiple rare variants in GPR10 in people with severe obesity and in normal weight controls. These variants impair ligand binding and G protein-dependent signalling in cells. Transgenic mice harbouring a loss of function GPR10 variant found in an individual with obesity, gain excessive weight due to decreased energy expenditure rather than increased food intake. This evidence supports a role for GPR10 in human energy homeostasis. Therapeutic targeting of GPR10 may represent an effective weight-loss strategy.


Subject(s)
Obesity , Receptors, G-Protein-Coupled , Animals , Humans , Mice , Energy Metabolism , Mice, Transgenic , Obesity/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Weight Gain/genetics
5.
Nat Med ; 28(12): 2537-2546, 2022 12.
Article in English | MEDLINE | ID: mdl-36536256

ABSTRACT

Serotonin reuptake inhibitors and receptor agonists are used to treat obesity, anxiety and depression. Here we studied the role of the serotonin 2C receptor (5-HT2CR) in weight regulation and behavior. Using exome sequencing of 2,548 people with severe obesity and 1,117 control individuals without obesity, we identified 13 rare variants in the gene encoding 5-HT2CR (HTR2C) in 19 unrelated people (3 males and 16 females). Eleven variants caused a loss of function in HEK293 cells. All people who carried variants had hyperphagia and some degree of maladaptive behavior. Knock-in male mice harboring a human loss-of-function HTR2C variant developed obesity and reduced social exploratory behavior; female mice heterozygous for the same variant showed similar deficits with reduced severity. Using the 5-HT2CR agonist lorcaserin, we found that depolarization of appetite-suppressing proopiomelanocortin neurons was impaired in knock-in mice. In conclusion, we demonstrate that 5-HT2CR is involved in the regulation of human appetite, weight and behavior. Our findings suggest that melanocortin receptor agonists might be effective in treating severe obesity in individuals carrying HTR2C variants. We suggest that HTR2C should be included in diagnostic gene panels for severe childhood-onset obesity.


Subject(s)
Obesity, Morbid , Receptor, Serotonin, 5-HT2C , Animals , Child , Female , Humans , Male , Mice , HEK293 Cells , Obesity/genetics , Receptor, Serotonin, 5-HT2C/genetics , Serotonin , Serotonin 5-HT2 Receptor Agonists/pharmacology , Adaptation, Psychological
6.
Front Endocrinol (Lausanne) ; 13: 891586, 2022.
Article in English | MEDLINE | ID: mdl-35846282

ABSTRACT

The intestinal hormone, glucose-dependent insulinotropic polypeptide (GIP), is involved in important physiological functions, including postprandial blood glucose homeostasis, bone remodeling, and lipid metabolism. While mutations leading to physiological changes can be identified in large-scale sequencing, no systematic investigation of GIP missense variants has been performed. Here, we identified 168 naturally occurring missense variants in the human GIP genes from three independent cohorts comprising ~720,000 individuals. We examined amino acid changing variants scattered across the pre-pro-GIP peptide using in silico effect predictions, which revealed that the sequence of the fully processed GIP hormone is more protected against mutations than the rest of the precursor protein. Thus, we observed a highly species-orthologous and population-specific conservation of the GIP peptide sequence, suggestive of evolutionary constraints to preserve the GIP peptide sequence. Elucidating the mutational landscape of GIP variants and how they affect the structural and functional architecture of GIP can aid future biological characterization and clinical translation.


Subject(s)
Blood Glucose , Incretins/metabolism , Receptors, G-Protein-Coupled , Amino Acid Sequence , Humans , Mutation, Missense , Selection, Genetic
7.
N Engl J Med ; 385(17): 1581-1592, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34614324

ABSTRACT

BACKGROUND: GNAS encodes the Gαs (stimulatory G-protein alpha subunit) protein, which mediates G protein-coupled receptor (GPCR) signaling. GNAS mutations cause developmental delay, short stature, and skeletal abnormalities in a syndrome called Albright's hereditary osteodystrophy. Because of imprinting, mutations on the maternal allele also cause obesity and hormone resistance (pseudohypoparathyroidism). METHODS: We performed exome sequencing and targeted resequencing in 2548 children who presented with severe obesity, and we unexpectedly identified 22 GNAS mutation carriers. We investigated whether the effect of GNAS mutations on melanocortin 4 receptor (MC4R) signaling explains the obesity and whether the variable clinical spectrum in patients might be explained by the results of molecular assays. RESULTS: Almost all GNAS mutations impaired MC4R signaling. A total of 6 of 11 patients who were 12 to 18 years of age had reduced growth. In these patients, mutations disrupted growth hormone-releasing hormone receptor signaling, but growth was unaffected in carriers of mutations that did not affect this signaling pathway (mean standard-deviation score for height, -0.90 vs. 0.75, respectively; P = 0.02). Only 1 of 10 patients who reached final height before or during the study had short stature. GNAS mutations that impaired thyrotropin receptor signaling were associated with developmental delay and with higher thyrotropin levels (mean [±SD], 8.4±4.7 mIU per liter) than those in 340 severely obese children who did not have GNAS mutations (3.9±2.6 mIU per liter; P = 0.004). CONCLUSIONS: Because pathogenic mutations may manifest with obesity alone, screening of children with severe obesity for GNAS deficiency may allow early diagnosis, improving clinical outcomes, and melanocortin agonists may aid in weight loss. GNAS mutations that are identified by means of unbiased genetic testing differentially affect GPCR signaling pathways that contribute to clinical heterogeneity. Monogenic diseases are clinically more variable than their classic descriptions suggest. (Funded by Wellcome and others.).


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/genetics , Mutation , Pediatric Obesity/genetics , Receptor, Melanocortin, Type 4/metabolism , Adolescent , Body Height , Child , Chromogranins/genetics , Female , GTP-Binding Protein alpha Subunits, Gs/deficiency , Humans , Male , Mutation, Missense , Receptors, Thyrotropin/metabolism , Signal Transduction , Exome Sequencing
8.
Nat Med ; 27(6): 1088-1096, 2021 06.
Article in English | MEDLINE | ID: mdl-34045736

ABSTRACT

Mutations in the melanocortin 4 receptor gene (MC4R) are associated with obesity but little is known about the prevalence and impact of such mutations throughout human growth and development. We examined the MC4R coding sequence in 5,724 participants from the Avon Longitudinal Study of Parents and Children, functionally characterized all nonsynonymous MC4R variants and examined their association with anthropometric phenotypes from childhood to early adulthood. The frequency of heterozygous loss-of-function (LoF) mutations in MC4R was ~1 in 337 (0.30%), considerably higher than previous estimates. At age 18 years, mean differences in body weight, body mass index and fat mass between carriers and noncarriers of LoF mutations were 17.76 kg (95% CI 9.41, 26.10), 4.84 kg m-2 (95% CI 2.19, 7.49) and 14.78 kg (95% CI 8.56, 20.99), respectively. MC4R LoF mutations may be more common than previously reported and carriers of such variants may enter adult life with a substantial burden of excess adiposity.


Subject(s)
Body Weight/genetics , Genetic Predisposition to Disease , Obesity/genetics , Receptor, Melanocortin, Type 4/genetics , Adolescent , Adult , Child , Child, Preschool , Heterozygote , Humans , Infant , Infant, Newborn , Loss of Function Mutation/genetics , Male , Obesity/epidemiology , Obesity/pathology , Phenotype , United Kingdom , Young Adult
9.
Cell Rep ; 34(12): 108862, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33761344

ABSTRACT

The Melanocortin-4 Receptor (MC4R) plays a pivotal role in energy homeostasis. We used human MC4R mutations associated with an increased or decreased risk of obesity to dissect mechanisms that regulate MC4R function. Most obesity-associated mutations impair trafficking to the plasma membrane (PM), whereas obesity-protecting mutations either accelerate recycling to the PM or decrease internalization, resulting in enhanced signaling. MC4R mutations that do not affect canonical Gαs protein-mediated signaling, previously considered to be non-pathogenic, nonetheless disrupt agonist-induced internalization, ß-arrestin recruitment, and/or coupling to Gαs, establishing their causal role in severe obesity. Structural mapping reveals ligand-accessible sites by which MC4R couples to effectors and residues involved in the homodimerization of MC4R, which is disrupted by multiple obesity-associated mutations. Human genetic studies reveal that endocytosis, intracellular trafficking, and homodimerization regulate MC4R function to a level that is physiologically relevant, supporting the development of chaperones, agonists, and allosteric modulators of MC4R for weight loss therapy.


Subject(s)
Body Weight/genetics , Endocytosis , Genetic Variation , Protein Multimerization , Receptor, Melanocortin, Type 4/genetics , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gs , HEK293 Cells , Humans , Models, Biological , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation , Receptor, Melanocortin, Type 4/chemistry , Signal Transduction , beta-Arrestins/metabolism
10.
Cell ; 177(3): 597-607.e9, 2019 04 18.
Article in English | MEDLINE | ID: mdl-31002796

ABSTRACT

The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor whose disruption causes obesity. We functionally characterized 61 MC4R variants identified in 0.5 million people from UK Biobank and examined their associations with body mass index (BMI) and obesity-related cardiometabolic diseases. We found that the maximal efficacy of ß-arrestin recruitment to MC4R, rather than canonical Gαs-mediated cyclic adenosine-monophosphate production, explained 88% of the variance in the association of MC4R variants with BMI. While most MC4R variants caused loss of function, a subset caused gain of function; these variants were associated with significantly lower BMI and lower odds of obesity, type 2 diabetes, and coronary artery disease. Protective associations were driven by MC4R variants exhibiting signaling bias toward ß-arrestin recruitment and increased mitogen-activated protein kinase pathway activation. Harnessing ß-arrestin-biased MC4R signaling may represent an effective strategy for weight loss and the treatment of obesity-related cardiometabolic diseases.


Subject(s)
Gain of Function Mutation/genetics , Obesity/pathology , Receptor, Melanocortin, Type 4/genetics , Signal Transduction , Adult , Aged , Body Mass Index , Coronary Artery Disease/complications , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Cyclic AMP/metabolism , Databases, Factual , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Obesity/complications , Obesity/metabolism , Polymorphism, Single Nucleotide , Receptor, Melanocortin, Type 4/chemistry , Receptor, Melanocortin, Type 4/metabolism , beta-Arrestins/metabolism
11.
Mol Metab ; 19: 49-64, 2019 01.
Article in English | MEDLINE | ID: mdl-30472415

ABSTRACT

OBJECTIVES: GPR142, which is highly expressed in pancreatic islets, has recently been deorphanized as a receptor for aromatic amino acids; however, its physiological role and pharmacological potential is unclear. METHODS AND RESULTS: We find that GPR142 is expressed not only in ß- but also in α-cells of the islets as well as in enteroendocrine cells, and we confirm that GPR142 is a highly selective sensor of essential aromatic amino acids, in particular Trp and oligopeptides with N-terminal Trp. GPR142 knock-out mice displayed a very limited metabolic phenotype but demonstrated that L-Trp induced secretion of pancreatic and gut hormones is mediated through GPR142 but that the receptor is not required for protein-induced hormone secretion. A synthetic GPR142 agonist stimulated insulin and glucagon as well as GIP, CCK, and GLP-1 secretion. In particular, GIP secretion was sensitive to oral administration of the GPR142 agonist an effect which in contrast to the other hormones was blocked by protein load. Oral administration of the GPR142 agonist increased [3H]-2-deoxyglucose uptake in muscle and fat depots mediated through insulin action while it lowered liver glycogen conceivably mediated through glucagon, and, consequently, it did not lower total blood glucose. Nevertheless, acute administration of the GPR142 agonist strongly improved oral glucose tolerance in both lean and obese mice as well as Zucker fatty rat. Six weeks in-feed chronic treatment with the GPR142 agonist did not affect body weight in DIO mice, but increased energy expenditure and carbohydrate utilization, lowered basal glucose, and improved insulin sensitivity. CONCLUSIONS: GPR142 functions as a sensor of aromatic amino acids, controlling GIP but also CCK and GLP-1 as well as insulin and glucagon in the pancreas. GPR142 agonists could have novel interesting potential in modifying metabolism through a balanced action of gut hormones as well as both insulin and glucagon.


Subject(s)
Islets of Langerhans/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acids, Aromatic/metabolism , Animals , Blood Glucose/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Rats , Rats, Zucker , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Tryptophan/metabolism
12.
Nat Med ; 24(5): 551-555, 2018 05.
Article in English | MEDLINE | ID: mdl-29736023

ABSTRACT

Genetic defects underlying the melanocortin-4 receptor (MC4R) signaling pathway lead to severe obesity. Three severely obese LEPR-deficient individuals were administered the MC4R agonist setmelanotide, resulting in substantial and durable reductions in hyperphagia and body weight over an observation period of 45-61 weeks. Compared to formerly developed and tested MC4R agonists, setmelanotide has the unique capability of activating nuclear factor of activated T cell (NFAT) signaling and restoring function of this signaling pathway for selected MC4R variants. Our data demonstrate the potency of setmelanotide in treatment of individuals with diverse MC4R-related pathway deficiencies.


Subject(s)
Receptor, Melanocortin, Type 4/agonists , Receptors, Leptin/deficiency , Weight Loss , Adolescent , Enzyme Activation/drug effects , HEK293 Cells , Humans , Male , Peptides/pharmacology , Receptors, Leptin/genetics , Type C Phospholipases/metabolism , Weight Loss/drug effects , Young Adult , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
13.
Cell Metab ; 27(2): 419-427.e4, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29414687

ABSTRACT

Palmitic acid hydroxystearic acids (PAHSAs) are endogenous lipids with anti-diabetic and anti-inflammatory effects. PAHSA levels are reduced in serum and adipose tissue of insulin-resistant people and high-fat diet (HFD)-fed mice. Here, we investigated whether chronic PAHSA treatment enhances insulin sensitivity and which receptors mediate PAHSA effects. Chronic PAHSA administration in chow- and HFD-fed mice raises serum and tissue PAHSA levels ∼1.4- to 3-fold. This improves insulin sensitivity and glucose tolerance without altering body weight. PAHSA administration in chow-fed, but not HFD-fed, mice augments insulin and glucagon-like peptide (GLP-1) secretion. PAHSAs are selective agonists for GPR40, increasing Ca+2 flux, but not intracellular cyclic AMP. Blocking GPR40 reverses improvements in glucose tolerance and insulin sensitivity in PAHSA-treated chow- and HFD-fed mice and directly inhibits PAHSA augmentation of glucose-stimulated insulin secretion in human islets. In contrast, GLP-1 receptor blockade in PAHSA-treated chow-fed mice reduces PAHSA effects on glucose tolerance, but not on insulin sensitivity. Thus, PAHSAs activate GPR40, which is involved in their beneficial metabolic effects.


Subject(s)
Glucose/metabolism , Homeostasis , Palmitic Acid/pharmacology , Receptors, G-Protein-Coupled/metabolism , Stearic Acids/pharmacology , Adiposity/drug effects , Animals , Eating/drug effects , HEK293 Cells , Homeostasis/drug effects , Humans , Inflammation/pathology , Insulin Resistance , Mice, Inbred C57BL
14.
Mol Metab ; 6(10): 1321-1329, 2017 10.
Article in English | MEDLINE | ID: mdl-29031731

ABSTRACT

OBJECTIVE: Pro-opiomelanocortin (POMC)-derived peptides act on neurons expressing the Melanocortin 4 receptor (MC4R) to reduce body weight. Setmelanotide is a highly potent MC4R agonist that leads to weight loss in diet-induced obese animals and in obese individuals with complete POMC deficiency. While POMC deficiency is very rare, 1-5% of severely obese individuals harbor heterozygous mutations in MC4R. We sought to assess the efficacy of Setmelanotide in human MC4R deficiency. METHODS: We studied the effects of Setmelanotide on mutant MC4Rs in cells and the weight loss response to Setmelanotide administration in rodent studies and a human clinical trial. We annotated the functional status of 369 published MC4R variants. RESULTS: In cells, we showed that Setmelanotide is significantly more potent at MC4R than the endogenous ligand alpha-melanocyte stimulating hormone and can disproportionally rescue signaling by a subset of severely impaired MC4R mutants. Wild-type rodents appear more sensitive to Setmelanotide when compared to MC4R heterozygous deficient mice, while MC4R knockout mice fail to respond. In a 28-day Phase 1b clinical trial, Setmelanotide led to weight loss in obese MC4R variant carriers. Patients with POMC defects upstream of MC4R show significantly more weight loss with Setmelanotide than MC4R deficient patients or obese controls. CONCLUSIONS: Setmelanotide led to weight loss in obese people with MC4R deficiency; however, further studies are justified to establish whether Setmelanotide can elicit clinically meaningful weight loss in a subset of the MC4R deficient obese population.


Subject(s)
Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/deficiency , alpha-MSH/analogs & derivatives , Adrenal Insufficiency/drug therapy , Adrenal Insufficiency/metabolism , Adult , Amino Acid Sequence , Animals , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Middle Aged , Obesity/drug therapy , Obesity/metabolism , Pro-Opiomelanocortin/deficiency , Pro-Opiomelanocortin/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/pharmacology
15.
Neurochem Int ; 102: 105-113, 2017 01.
Article in English | MEDLINE | ID: mdl-27916541

ABSTRACT

GPR139 is an orphan G protein-coupled receptor that is expressed primarily in the brain. Not much is known regarding the function of GPR139. Recently we have shown that GPR139 is activated by the amino acids l-tryptophan and l-phenylalanine (EC50 values of 220 µM and 320 µM, respectively), as well as di-peptides comprised of aromatic amino acids. This led us to hypothesize that GPR139 may be activated by peptides. Sequence alignment of the binding cavities of all class A GPCRs, revealed that the binding pocket of the melanocortin 4 receptor is similar to that of GPR139. Based on the chemogenomics principle "similar targets bind similar ligands", we tested three known endogenous melanocortin 4 receptor agonists; adrenocorticotropic hormone (ACTH) and α- and ß-melanocyte stimulating hormone (α-MSH and ß-MSH) on CHO-k1 cells stably expressing the human GPR139 in a Fluo-4 Ca2+-assay. All three peptides, as well as their conserved core motif HFRW, were found to activate GPR139 in the low micromolar range. Moreover, we found that peptides consisting of nine or ten N-terminal residues of α-MSH activate GPR139 in the submicromolar range. α-MSH1-9 was found to correspond to the product of a predicted cleavage site in the pre-pro-protein pro-opiomelanocortin (POMC). Our results demonstrate that GPR139 is a peptide receptor, activated by ACTH, α-MSH, ß-MSH, the conserved core motif HFRW as well as a potential endogenous peptide α-MSH1-9. Further studies are needed to determine the functional relevance of GPR139 mediated signaling by these peptides.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Melanocytes/metabolism , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , alpha-MSH/metabolism , beta-MSH/metabolism , Amino Acid Motifs , Animals , CHO Cells , Cricetulus , Melanocyte-Stimulating Hormones/metabolism , Pro-Opiomelanocortin/metabolism , Receptor, Melanocortin, Type 4/metabolism
16.
J Biol Chem ; 290(40): 24495-508, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26269596

ABSTRACT

X-ray structures, molecular dynamics simulations, and mutational analysis have previously indicated that an extended water hydrogen bond network between trans-membranes I-III, VI, and VII constitutes an allosteric interface essential for stabilizing different active and inactive helical constellations during the seven-trans-membrane receptor activation. The neurokinin-1 receptor signals efficiently through Gq, Gs, and ß-arrestin when stimulated by substance P, but it lacks any sign of constitutive activity. In the water hydrogen bond network the neurokinin-1 has a unique Glu residue instead of the highly conserved AspII:10 (2.50). Here, we find that this GluII:10 occupies the space of a putative allosteric modulating Na(+) ion and makes direct inter-helical interactions in particular with SerIII:15 (3.39) and AsnVII:16 (7.49) of the NPXXY motif. Mutational changes in the interface between GluII:10 and AsnVII:16 created receptors that selectively signaled through the following: 1) Gq only; 2) ß-arrestin only; and 3) Gq and ß-arrestin but not through Gs. Interestingly, increased constitutive Gs but not Gq signaling was observed by Ala substitution of four out of the six core polar residues of the network, in particular SerIII:15. Three residues were essential for all three signaling pathways, i.e. the water-gating micro-switch residues TrpVI:13 (6.48) of the CWXP motif and TyrVII:20 (7.53) of the NPXXY motif plus the totally conserved AsnI:18 (1.50) stabilizing the kink in trans-membrane VII. It is concluded that the interface between position II:10 (2.50), III:15 (3.39), and VII:16 (7.49) in the center of the water hydrogen bond network constitutes a focal point for fine-tuning seven trans-membrane receptor conformations activating different signal transduction pathways.


Subject(s)
Arrestins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neurokinin-1/metabolism , Alanine/chemistry , Allosteric Site , Animals , COS Cells , Chlorocebus aethiops , Crystallography, X-Ray , DNA Mutational Analysis , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Monte Carlo Method , Protein Conformation , Receptors, Ghrelin/metabolism , Signal Transduction , Sodium/chemistry , Transfection , Water/chemistry , beta-Arrestins
17.
J Biol Chem ; 287(40): 33488-502, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22846991

ABSTRACT

Based on a rare, natural Glu for Ala-204(C+6) variant located six residues after the conserved Cys residue in extracellular loop 2b (ECL2b) associated with selective elimination of the high constitutive signaling of the ghrelin receptor, this loop was subjected to a detailed structure functional analysis. Introduction of Glu in different positions demonstrated that although the constitutive signaling was partly reduced when introduced in position 205(C+7) it was only totally eliminated in position 204(C+6). No charge-charge interaction partner could be identified for the Glu(C+6) variant despite mutational analysis of a number of potential partners in the extracellular loops and outer parts of the transmembrane segments. Systematic probing of position 204(C+6) with amino acid residues of different physicochemical properties indicated that a positively charged Lys surprisingly provided phenotypes similar to those of the negatively charged Glu residue. Computational chemistry analysis indicated that the propensity for the C-terminal segment of extracellular loop 2b to form an extended α-helix was increased from 15% in the wild type to 89 and 82% by introduction in position 204(C+6) of a Glu or a Lys residue, respectively. Moreover, the constitutive activity of the receptor was inhibited by Zn(2+) binding in an engineered metal ion site, stabilizing an α-helical conformation of this loop segment. It is concluded that the high constitutive activity of the ghrelin receptor is dependent upon flexibility in the C-terminal segment of extracellular loop 2 and that mutations or ligand binding that constrains this segment and thereby conceivably the movements of transmembrane domain V relative to transmembrane domain III inhibits the high constitutive signaling.


Subject(s)
Receptors, Ghrelin/metabolism , Alanine/chemistry , Amino Acid Sequence , Animals , Arrestins/metabolism , COS Cells , Chlorocebus aethiops , DNA Mutational Analysis , HEK293 Cells , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Receptors, Ghrelin/chemistry , Signal Transduction , beta-Arrestins
18.
J Med Chem ; 55(17): 7437-49, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22920150

ABSTRACT

The ghrelin receptor displays a high constitutive activity suggested to be involved in the regulation of appetite and food intake. Here, we have created peptides with small changes in the core binding motif -wFw- of the hexapeptide KwFwLL-NH(2) that can swap the peptide behavior from inverse agonism to agonism, indicating the importance of this sequence. Introduction of ß-(3-benzothienyl)-d-alanine (d-Bth), 3,3-diphenyl-d-alanine (d-Dip) and 1-naphthyl-d-alanine (d-1-Nal) at position 2 resulted in highly potent and efficient inverse agonists, whereas the substitution of d-tryptophane at position 4 with 1-naphthyl-d-alanine (d-1-Nal) and 2-naphthyl-d-alanine (d-2-Nal) induces agonism in functional assays. Competitive binding studies showed a high affinity of the inverse agonist K-(d-1-Nal)-FwLL-NH(2) at the ghrelin receptor. Moreover, mutagenesis studies of the receptor revealed key positions for the switch between inverse agonist and agonist response. Hence, only minor changes in the peptide sequence can decide between agonism and inverse agonism and have a major impact on the biological activity.


Subject(s)
Receptors, Ghrelin/drug effects , Animals , COS Cells , Chlorocebus aethiops , Feeding Behavior/drug effects , Humans , Models, Molecular , Mutagenesis , Oligopeptides/chemistry , Oligopeptides/pharmacology , Rats , Receptors, Ghrelin/agonists , Receptors, Ghrelin/antagonists & inhibitors
19.
Oncology ; 81(3-4): 243-50, 2011.
Article in English | MEDLINE | ID: mdl-22116456

ABSTRACT

OBJECTIVE: Alterations in the transforming growth factor-ß (TGF-ß) signaling cascade are engaged in the development of human neoplasms through the deregulation of proliferation, differentiation and migration. However, in endometrial cancer, the role of endoglin, which acts as an accessory receptor in the TGF-ß pathway, is still unknown. The aim of our study was the evaluation of endoglin mRNA and protein expression levels in endometrial cancer as compared to normal endometrium. TGF-ß(1) and TGF-ß type II receptor were involved in the investigation since they directly cooperate with endoglin during signal propagation. Obtained results were correlated with clinicopathological parameters of studied material to determine endoglin contribution to tumor development and progression. METHODS: mRNA level assessment was performed using real-time technique, whereas protein expression was determined by ELISA assay. RESULTS: The endoglin mRNA level was not significantly altered in cancerous samples as compared to normal tissue, whereas its protein level demonstrated significant upregulation (p < 0.001) associated with increased tumor malignancy, assessed by histological grade and myometrium infiltration. CONCLUSIONS: An increase in endoglin protein expression level may interfere with the oncogenic potential of TGF-ß(1) and TGF-ß type II receptor in endometrial cancer. Correlation of the endoglin level with pronounced cancer malignancy suggests that it may be regarded as a potential prognostic marker of primary endometrial cancer.


Subject(s)
Antigens, CD/biosynthesis , Endometrial Neoplasms/metabolism , Receptors, Cell Surface/biosynthesis , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Down-Regulation , Endoglin , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Cell Surface/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Up-Regulation
20.
PLoS One ; 6(9): e24644, 2011.
Article in English | MEDLINE | ID: mdl-21931793

ABSTRACT

BACKGROUND: The melanocortin 1 receptor (MC1R) constitutes a key regulator of melanism. Consequently, many naturally-occurring MC1R mutations are associated with a change in color. An example is the Glu-to-Lys substitution found at position II:20/2.60 in the top of transmembrane helix II which has been identified in melanic mice and several other species. This mutation induces a pronounced increase in MC1R constitutive activity suggesting a link between constitutive activity and melanism which is corroborated by the attenuation of α-melanocyte stimulating hormone (αMSH) induced activation. However, the mechanism by which the mutation induces constitutive activity is currently not known. METHODOLOGY/PRINCIPAL FINDINGS: Here we characterize the constitutive activity, cell surface expression and internalization of the mouse mutant, Mc1r E92K. As previously reported, only positively charged residues at position II:20/2.60 induced an increase in constitutive activity as measured by cAMP accumulation and CREB activation. Furthermore, the mutation induced a constitutive recruitment of ß-arrestin. This phenomenon is only observed in MC1R, however, as the equivalent mutations in MC2-5R had no effect on receptor signaling. Interestingly, the mutation did not induce constitutive ERK1/2 phosphorylation or increase the internalization rate indicating the constitutive activity to be biased. Finally, to identify regions of importance for the increased constitutive activity of Mc1r E92K, we employed a chimeric approach and identified G102 and L110 in the extracellular loop 1 to be selectively important for the constitutive activity as this, but not αMSH-mediated activation, was abolished upon Ala substitution. CONCLUSIONS/SIGNIFICANCE: It is concluded that the E92K mutation induces an active conformation distinct from that induced by αMSH and that the extracellular loop 1 is involved in maintaining this conformational state. In turn, the results suggest that in MC1R, which lacks an extracellular loop 2, the first extracellular loop may play a more prominent role during receptor activation than in general.


Subject(s)
Arrestin/metabolism , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptor, Melanocortin, Type 1/metabolism , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Microscopy, Confocal , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Receptor, Melanocortin, Type 1/genetics , Signal Transduction/genetics , Signal Transduction/physiology , alpha-MSH/genetics , alpha-MSH/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...