Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Chem Biol ; 30(8): 933-942.e6, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37453421

ABSTRACT

Insulin resistance (IR) is the root cause of type II diabetes, yet no safe treatment is available to address it. Using a high throughput compatible assay that measures real-time translocation of the glucose transporter glucose transporter 4 (GLUT4), we identified small molecules that potentiate insulin action. In vivo, these insulin sensitizers improve insulin-stimulated GLUT4 translocation, glucose tolerance, and glucose uptake in a model of IR. Using proteomic and CRISPR-based approaches, we identified the targets of those compounds as Unc119 proteins and solved the structure of Unc119 bound to the insulin sensitizer. This study identifies compounds that have the potential to be developed into diabetes treatment and establishes Unc119 proteins as targets for improving insulin sensitivity.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Insulin/metabolism , Diabetes Mellitus, Type 2/drug therapy , Proteomics , Glucose/metabolism , Protein Transport , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 4/metabolism
2.
J Biol Chem ; 299(1): 102814, 2023 01.
Article in English | MEDLINE | ID: mdl-36539034

ABSTRACT

Energy homeostasis is a complex system involving multiple hormones, neuropeptides, and receptors. Prokineticins (PK1 and PK2) are agonists to two G protein-coupled receptors, prokineticin receptor 1 and 2 (PKR1 and PKR2), which decrease food intake when injected in rodents. The relative contribution of PKR1 and PKR2 to the anorexigenic effect of PK2 and their site of action in the brain have not yet been elucidated. While PKR1 and PKR2 are both expressed in the hypothalamus, a central region involved in the control of energy homeostasis, PKR2 is also present in the amygdala, which has recently been shown to regulate food intake in response to several anorexigenic signals. PKR trafficking and signaling are inhibited by the melanocortin receptor accessory protein 2 (MRAP2), thus suggesting that MRAP2 has the potential to alter the anorexigenic activity of PK2 in vivo. In this study, we investigated the importance of PKR1 and PKR2 for PK2-mediated inhibition of food intake, the brain region involved in this function, and the effect of MRAP2 on PK2 action in vivo. Using targeted silencing of PKR2 and chemogenetic manipulation of PKR2 neurons, we show that the anorexigenic effect of PK2 is mediated by PKR2 in the amygdala and that altering MRAP2 expression in PKR2 neurons modulates the activity of PK2. Collectively, our results provide evidence that inhibition of food intake by PKs is not mediated through activation of hypothalamic neurons but rather amygdala PKR2 neurons and further establishes the importance of MRAP2 in the regulation of energy homeostasis.


Subject(s)
Gastrointestinal Hormones , Neuropeptides , Carrier Proteins/metabolism , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Gastrointestinal Hormones/pharmacology , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
3.
J Biol Chem ; 298(6): 102057, 2022 06.
Article in English | MEDLINE | ID: mdl-35605660

ABSTRACT

The melanocortin receptor accessory protein 2 (MRAP2) is essential for several physiological functions of the ghrelin receptor growth hormone secretagogue receptor 1a (GHSR1a), including increasing appetite and suppressing insulin secretion. In the absence of MRAP2, GHSR1a displays high constitutive activity and a weak G-protein-mediated response to ghrelin and readily recruits ß-arrestin. In the presence of MRAP2, however, G-protein-mediated signaling via GHSR1a is strongly dependent on ghrelin stimulation and the recruitment of ß-arrestin is significantly diminished. To better understand how MRAP2 modifies GHSR1a signaling, here we investigated the role of several phosphorylation sites within the C-terminal tail and third intracellular loop of GHSR1a, as well as the mechanism behind MRAP2-mediated inhibition of ß-arrestin recruitment. We show that Ser252 and Thr261 in the third intracellular loop of GHSR1a contribute to ß-arrestin recruitment, whereas the C-terminal region is not essential for ß-arrestin interaction. Additionally, we found that MRAP2 inhibits GHSR1a phosphorylation by blocking the interaction of GRK2 and PKC with the receptor. Taken together, these data suggest that MRAP2 alters GHSR1a signaling by directly impacting the phosphorylation state of the receptor and that the C-terminal tail of GHSR1a prevents rather than contribute to ß-arrestin recruitment.


Subject(s)
Adaptor Proteins, Signal Transducing , Ghrelin , Receptors, Ghrelin , beta-Arrestins , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , GTP-Binding Proteins/metabolism , Ghrelin/metabolism , Melanocortins , Phosphorylation , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , beta-Arrestins/genetics , beta-Arrestins/metabolism
4.
Front Neurosci ; 15: 730417, 2021.
Article in English | MEDLINE | ID: mdl-34483834

ABSTRACT

Prokineticin receptors are GPCRs involved in several physiological processes including the regulation of energy homeostasis, nociception, and reproductive function. PKRs are inhibited by the endogenous accessory protein MRAP2 which prevents them from trafficking to the plasma membrane. Very little is known about the importance of post-translational modification of PKRs and their role in receptor trafficking and signaling. Here we identify 2 N-linked glycosylation sites within the N-terminal region of PKR2 and demonstrate that glycosylation of PKR2 at position 27 is important for its plasma membrane localization and signaling. Additionally, we show that glycosylation at position 7 results in a decrease in PKR2 signaling through Gαs without impairing Gαq/ 11 signaling.

5.
Gastroenterology ; 161(1): 225-238.e15, 2021 07.
Article in English | MEDLINE | ID: mdl-33753103

ABSTRACT

BACKGROUND & AIMS: Tumor-infiltrating neutrophils (polymorphonuclear neutrophils [PMNs]) are a prominent feature of colorectal cancer (CRC), where they can promote cytotoxicity or exacerbate disease outcomes. We recently showed that in acute colon injury, PMNs can increase DNA double-strand break (DSB) burden and promote genomic instability via microRNA-dependent inhibition of homologous recombination (HR) repair. In this study, we aimed to establish whether in inflamed colon, neutrophils shape the DSB-repair responses to impact CRC progression and sensitivity/resistance to DNA-repair targeted therapy. METHODS: Human sporadic CRC biopsies, The Cancer Genome Atlas gene expression analyses, tumor xenografts, and murine CRC models, as well as small-molecule inhibition of key DSB-repair factors were leveraged to investigate changes in the DSB-repair landscape and identify unique CRC responses with/without tumor infiltration by PMNs. RESULTS: We reveal that neutrophils exert a functional dualism in cancer cells, driving temporal modulation of the DNA damage landscape and resolution of DSBs. PMNs were found to promote HR deficiency in low-grade CRC by miR-155-dependent downregulation of RAD51, thus attenuating tumor growth. However, neutrophil-mediated genotoxicity due to accumulation of DSBs led to the induction of non-homologous end-joining (NHEJ), allowing for survival and growth of advanced CRC. Our findings identified a PMN-induced HR-deficient CRC phenotype, featuring low RAD51 and low Ku70 levels, rendering it susceptible to synthetic lethality induced by clinically approved PARP1 inhibitor Olaparib. We further identified a distinct PMN-induced HR-deficient CRC phenotype, featuring high Ku70 and heightened NHEJ, which can be therapeutically targeted by specific inhibition of NHEJ. CONCLUSIONS: Our work delineates 2 mechanism-based translatable therapeutic interventions in sporadic CRC.


Subject(s)
Colitis-Associated Neoplasms/immunology , Colorectal Neoplasms/immunology , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Neutrophils/immunology , Tumor Microenvironment/immunology , Animals , Coculture Techniques , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Databases, Genetic , HCT116 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Neutrophils/metabolism , Phenotype , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Xenograft Model Antitumor Assays
6.
iScience ; 23(6): 101216, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32535024

ABSTRACT

Ghrelin regulates both energy intake and glucose homeostasis. In the endocrine pancreas, ghrelin inhibits insulin release to prevent hypoglycemia during fasting. The mechanism through which this is accomplished is unclear, but recent studies suggest that ghrelin acts on δ cells to stimulate somatostatin release, which in turn inhibits insulin release from ß cells. Recently, the Melanocortin Receptor Accessory Protein 2 (MRAP2) was identified as an essential partner of the ghrelin receptor (GHSR1a) in mediating the central orexigenic action of ghrelin. In this study we show that MRAP2 is expressed in islet δ cells and is required for ghrelin to elicit a calcium response in those cells. Additionally, we show that both global and δ cell targeted deletion of MRAP2 abrogates the insulinostatic effect of ghrelin. Together, these findings establish that ghrelin signaling within δ cells is essential for the inhibition of insulin release and identify MRAP2 as a regulator of insulin secretion.

7.
Sci Signal ; 13(613)2020 01 07.
Article in English | MEDLINE | ID: mdl-31911434

ABSTRACT

Ghrelin is a hormone secreted by the stomach during fasting periods and acts through its receptor, the growth hormone secretagogue 1a (GHSR1a), to promote food intake and prevent hypoglycemia. As such, GHSR1a is an important regulator of energy and glucose homeostasis and a target for the treatment of obesity. Here, we showed that the accessory protein MRAP2 altered GHSR1a signaling by inhibiting its constitutive activity, as well as by enhancing its G protein-dependent signaling and blocking the recruitment and signaling of ß-arrestin in response to ghrelin. In addition, the effects of MRAP2 on the Gαq and ß-arrestin pathways were independent and involved distinct regions of MRAP2. These findings may have implications for the regulation of ghrelin function in vivo and the role of MRAP2 in energy homeostasis. They also show that accessory proteins can bias signaling downstream of GPCRs in response to their endogenous agonist.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Energy Metabolism , Receptors, Ghrelin/immunology , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Animals , CHO Cells , Cricetulus , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Receptors, Ghrelin/genetics
8.
Hypertension ; 75(2): 569-579, 2020 02.
Article in English | MEDLINE | ID: mdl-31865781

ABSTRACT

Cascade-specific termination of G protein signaling is catalyzed by the RGS (regulator of G protein signaling) family members, including RGS2. Angiotensin, vasopressin, and endothelin are implicated in preeclampsia, and RGS2 is known to inhibit G protein cascades activated by these hormones. Mutations in RGS2 are associated with human hypertension and increased risk of developing preeclampsia and its sequelae. RGS family members are known to influence maternal vascular function, but the role of RGS2 within the placenta has not been explored. Here, we hypothesized that reduced expression of RGS2 within the placenta represents a risk factor for the development of preeclampsia. Although cAMP/CREB signaling was enriched in placentas from human pregnancies affected by preeclampsia compared with clinically matched controls and RGS2 is known to be a CREB-responsive gene, RGS2 mRNA was reduced in placentas from pregnancies affected by preeclampsia. Experimentally reducing Rgs2 expression within the feto-placental unit was sufficient to induce preeclampsia-like phenotypes in pregnant wild-type C57BL/6J mice. Stimulation of RGS2 transcription within immortalized human HTR8/SVneo trophoblasts by cAMP/CREB signaling was discovered to be dependent on the activity of histone deacetylase activity, and more specifically, HDAC9 (histone deacetylase-9), and HDAC9 expression was reduced in placentas from human pregnancies affected by preeclampsia. We conclude that reduced expression of RGS2 within the placenta may mechanistically contribute to preeclampsia. More generally, this work identifies RGS2 as an HDAC9-dependent CREB-responsive gene, which may contribute to reduced RGS2 expression in placenta during preeclampsia.


Subject(s)
Gene Expression Regulation , Placenta/metabolism , Pre-Eclampsia/genetics , Pregnancy, Animal , RGS Proteins/genetics , RNA, Messenger/biosynthesis , Animals , Disease Models, Animal , Disease Progression , Female , Humans , Mice , Mice, Inbred C57BL , Pre-Eclampsia/metabolism , Pregnancy , RGS Proteins/biosynthesis , RNA, Messenger/genetics , Signal Transduction
9.
Nat Commun ; 8(1): 713, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28959025

ABSTRACT

Ghrelin is the only known circulating orexigenic hormone. It is primarily secreted by the stomach and acts at its receptor, the growth hormone secretagogue receptor 1a (GHSR1a), in the hypothalamus to signal hunger and promote food intake. The melanocortin receptor accessory protein 2 (MRAP2) was previously shown to regulate energy homeostasis through the modulation of the activity of the melanocortin-4 receptor and prokineticin receptors. In this study we identify MRAP2 as a partner of ghrelin-GHSR1a signaling. We show that MRAP2 interacts with GHSR1a and potentiates ghrelin-stimulated signaling both in vitro and in vivo. We demonstrate that in the absence of MRAP2, fasting fails to activate agouti-related protein neurons. In addition, we show that the orexigenic effect of ghrelin is lost in mice lacking MRAP2. Our results suggest that MRAP2 is an important modulator of the energy homeostasis machinery that operates through the regulation of multiple GPCRs throughout the hypothalamus.Melanocortin receptor accessory protein 2 (MRAP2) is an adaptor protein that contributes to melanocortin-4 receptor and prokineticin receptor 1 signalling. Here the authors show that MRAP2 also regulates ghrelin receptor signalling in the hypothalamus and starvation sensing in mice.


Subject(s)
Hunger/physiology , Receptor Activity-Modifying Proteins/metabolism , Receptors, Ghrelin/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Eating , Energy Metabolism , Ghrelin/metabolism , Homeostasis , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor Activity-Modifying Proteins/genetics , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Receptors, Ghrelin/genetics
10.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2322-2329, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28939058

ABSTRACT

The Melanocortin Receptor Accessory Protein 2 (MRAP2) regulates the activity of several GPCRs involved in the control of food intake and energy expenditure. While MRAP2 was originally thought to exclusively interact with melanocortin receptors we have recently shown that it interacts with and inhibits the trafficking and signaling of the prokineticin receptor 1 (PKR1). In this study we demonstrate a new role of MRAP2 in the regulation of the orexin receptor 1 (OX1R) and identify the specific regions of MRAP2 required for the regulation of OX1R and PKR1. Importantly, like MC4R and PKRs, OX1R is predominately expressed in the brain where it regulates food intake. By demonstrating that MRAP2 modulates the activity of OX1R we further establish the critical role of MRAP2 in the control of energy homeostasis.


Subject(s)
Carrier Proteins/metabolism , Eating/genetics , Orexin Receptors/genetics , Receptors, G-Protein-Coupled/genetics , Adaptor Proteins, Signal Transducing , Animals , CHO Cells , Calcium Signaling/genetics , Carrier Proteins/genetics , Cricetulus , Energy Metabolism/genetics , Humans , Orexins/genetics , Orexins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism
11.
Biochim Biophys Acta Mol Basis Dis ; 1863(10 Pt A): 2462-2467, 2017 10.
Article in English | MEDLINE | ID: mdl-28499989

ABSTRACT

G-protein coupled receptors (GPCRs) are regulated by numerous proteins including kinases, G-proteins, ß-arrestins and accessory proteins. Several families of GPCR accessory proteins like Receptor Activity Modifying Proteins, Receptor Transporting Proteins and Melanocortin Receptor Accessory Proteins (MRAPs) have been identified as regulator of receptor trafficking, signaling and ligand specificity. The MRAP family contains two members, MRAP1 and MRAP2, responsible for the formation of a functional ACTH receptor and for the regulation of energy homeostasis respectively. Like all known GPCR accessory proteins, MRAPs are single transmembrane proteins, however, they form a unique structure since they assemble as an anti-parallel homodimer. Moreover, the accepted idea that MRAPs are specific regulators of melanocortin receptors was recently challenged by the discovery that MRAP2 inhibits the activity of prokineticin receptors. Recent studies are starting to explain the role of the unusual structure of MRAPs and to illustrate the importance of MRAP2 for the maintenance of both energy and glucose homeostasis. This article is part of a Special Issue entitled: Melanocortin Receptors - edited by Ya-Xiong Tao.


Subject(s)
Carrier Proteins/metabolism , Adaptor Proteins, Signal Transducing , Adrenal Glands/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Energy Metabolism , Glucose/metabolism , Humans , Melanocortins/metabolism , Protein Conformation , Protein Interaction Maps , Receptors, G-Protein-Coupled/metabolism , Receptors, Melanocortin/metabolism , Sequence Alignment
12.
Elife ; 52016 Feb 01.
Article in English | MEDLINE | ID: mdl-26829592

ABSTRACT

The Melanocortin Receptor Accessory Protein 2 (MRAP2) is an important regulator of energy homeostasis and its loss causes severe obesity in rodents. MRAP2 mediates its action in part through the potentiation of the MC4R, however, it is clear that MRAP2 is expressed in tissues that do not express MC4R, and that the deletion of MRAP2 does not recapitulate the phenotype of Mc4r KO mice. Consequently, we hypothesized that other GPCRs involved in the control of energy homeostasis are likely to be regulated by MRAP2. In this study we identified PKR1 as the first non-melanocortin GPCR to be regulated by MRAP2. We show that MRAP2 significantly and specifically inhibits PKR1 signaling. We also demonstrate that PKR1 and MRAP2 co-localize in neurons and that Mrap2 KO mice are hypersensitive to PKR1 stimulation. This study not only identifies new partners of MRAP2 but also a new pathway through which MRAP2 regulates energy homeostasis.


Subject(s)
Eating , Receptor Activity-Modifying Proteins/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Mice , Mice, Knockout , Neurons/chemistry
13.
Nature ; 520(7545): 94-8, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25600267

ABSTRACT

The regulated release of anorexigenic α-melanocyte stimulating hormone (α-MSH) and orexigenic Agouti-related protein (AgRP) from discrete hypothalamic arcuate neurons onto common target sites in the central nervous system has a fundamental role in the regulation of energy homeostasis. Both peptides bind with high affinity to the melanocortin-4 receptor (MC4R); existing data show that α-MSH is an agonist that couples the receptor to the Gαs signalling pathway, while AgRP binds competitively to block α-MSH binding and blocks the constitutive activity mediated by the ligand-mimetic amino-terminal domain of the receptor. Here we show that, in mice, regulation of firing activity of neurons from the paraventricular nucleus of the hypothalamus (PVN) by α-MSH and AgRP can be mediated independently of Gαs signalling by ligand-induced coupling of MC4R to closure of inwardly rectifying potassium channel, Kir7.1. Furthermore, AgRP is a biased agonist that hyperpolarizes neurons by binding to MC4R and opening Kir7.1, independently of its inhibition of α-MSH binding. Consequently, Kir7.1 signalling appears to be central to melanocortin-mediated regulation of energy homeostasis within the PVN. Coupling of MC4R to Kir7.1 may explain unusual aspects of the control of energy homeostasis by melanocortin signalling, including the gene dosage effect of MC4R and the sustained effects of AgRP on food intake.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Potassium Channels, Inwardly Rectifying/metabolism , Receptor, Melanocortin, Type 4/metabolism , Action Potentials , Agouti-Related Protein/metabolism , Animals , Eating/genetics , Energy Metabolism , Female , HEK293 Cells , Homeostasis/genetics , Humans , Ligands , Male , Melanocortins/metabolism , Mice , Receptor, Melanocortin, Type 4/genetics , Signal Transduction/genetics , alpha-MSH/metabolism
14.
Science ; 341(6143): 278-81, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23869017

ABSTRACT

The melanocortin-4 receptor (MC4R) is essential for control of energy homeostasis in vertebrates. MC4R interacts with melanocortin receptor accessory protein 2 (MRAP2) in vitro, but its functions in vivo are unknown. We found that MRAP2a, a larval form, stimulates growth of zebrafish by specifically blocking the action of MC4R. In cell culture, this protein binds MC4R and reduces the ability of the receptor to bind its ligand, α-melanocyte-stimulating hormone (α-MSH). A paralog, MRAP2b, expressed later in development, also binds MC4R but increases ligand sensitivity. Thus, MRAP2 proteins allow for developmental control of MC4R activity, with MRAP2a blocking its function and stimulating growth during larval development, whereas MRAP2b enhances responsiveness to α-MSH once the zebrafish begins feeding, thus increasing the capacity for regulated feeding and growth.


Subject(s)
Receptor Activity-Modifying Proteins/metabolism , Receptor, Melanocortin, Type 4/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Embryo, Nonmammalian/metabolism , Energy Metabolism , HEK293 Cells , Humans , Receptor Activity-Modifying Proteins/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , alpha-MSH/metabolism , alpha-MSH/pharmacology
15.
Med Sci (Paris) ; 28(10): 845-51, 2012 Oct.
Article in French | MEDLINE | ID: mdl-23067415

ABSTRACT

G-protein-coupled receptors (GPCR) are a major family of drug targets. Essentially all drugs targeting these receptors on the market compete with the endogenous ligand (agonists or antagonists) for binding the receptor. Recently, non-competitive compounds binding to distinct sites from the cognate ligand were documented in various classes of these receptors. These compounds, called allosteric modulators, generally endowed of a better selectivity are able to modulate specifically the endogenous signaling of the receptor. To better understand the promising potential of this class of GPCRs targeting compounds, this review highlights the properties of allosteric modulators, the strategies used to identify them and the challenges associated with the development of these compounds.


Subject(s)
Allosteric Site/physiology , Drug Design , Molecular Targeted Therapy , Receptors, G-Protein-Coupled , Allosteric Regulation , Animals , High-Throughput Screening Assays , Humans , Models, Biological , Molecular Targeted Therapy/methods , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism
16.
Gen Comp Endocrinol ; 174(1): 5-14, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21846469

ABSTRACT

Analysis of the functional expression of the melanocortin 2 receptor (MC2R) from a rather broad spectrum of vertebrates indicates that MC2R is exclusively selective for the ligand, ACTH, and the melanocortin receptor accessory protein 1 (MRAP1) is required for high affinity ACTH binding and activation of MC2R. A phylogenetic analysis of MRAP1 suggested that tetrapod sequences and bony fish sequences may represent two distinct trends in the evolution of the mrap1 gene. To test this hypothesis, a frog (Xenopus tropicalis) MC2R was expressed in CHO cells either in the presence of a tetrapod (mouse) MRAP1 or a bony fish (zebrafish) MRAP1. The response of frog MC2R to different concentrations of human ACTH(1-24) was more robust in the presence of mouse MRAP1 than in the presence of zebrafish MRAP1. Conversely, the cAMP response mediated by the rainbow trout (Oncorhynchus mykiss) MC2R was almost twofold higher and occurred at 1000-fold lower ACTH concentration in the presence of zebrafish MRAP1 than in the presence of mouse MRAP1. Collectively, these experiments raise the possibility that at least two distinct trends have emerged in the co-evolution of MC2R/MRAP1 interactions during the radiation of the vertebrates.


Subject(s)
Anura/metabolism , Oncorhynchus mykiss/metabolism , Receptor, Melanocortin, Type 2/metabolism , Adrenocorticotropic Hormone/metabolism , Animals , CHO Cells , Cricetinae , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Protein Binding , Receptor, Melanocortin, Type 2/genetics
17.
Eur J Pharmacol ; 660(1): 13-20, 2011 Jun 11.
Article in English | MEDLINE | ID: mdl-21211527

ABSTRACT

The melanocortin MC(3) receptor remains the most enigmatic of the melanocortin receptors with regard to its physiological functions. The receptor is expressed both in the CNS and in multiple tissues in the periphery. It appears to be an inhibitory autoreceptor on proopiomelanocortin neurons, yet global deletion of the receptor causes an obesity syndrome. Knockout of the receptor increases adipose mass without a readily measurable increase in food intake or decrease in energy expenditure. And finally, no melanocortin MC(3) receptor null humans have been identified and associations between variant alleles of the melanocortin MC(3) receptor and diseases remain controversial, so the physiological role of the receptor in humans remains to be determined.


Subject(s)
Receptor, Melanocortin, Type 3/metabolism , Amino Acid Sequence , Animals , Autoreceptors/chemistry , Autoreceptors/genetics , Autoreceptors/immunology , Autoreceptors/metabolism , Gene Expression Regulation , Humans , Molecular Sequence Data , Receptor, Melanocortin, Type 3/chemistry , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 3/immunology
18.
Eur J Pharmacol ; 660(1): 94-102, 2011 Jun 11.
Article in English | MEDLINE | ID: mdl-21211532

ABSTRACT

The melanocortin 2 (MC(2)) receptor differs from other melanocortin family members in its pharmacological profile and reliance on an accessory protein, MC(2) receptor accessory protein (MRAP), for surface expression and signal transduction. To identify features of the MC(2) receptor responsible for these characteristics, we created chimeras between MC(2) and MC(4) receptors and expressed these in CHO cells, where MRAP is essential for trafficking and signaling by MC(2) but not MC(4) receptors. Replacing the first transmembrane segment of the MC(2) receptor with the corresponding region from the MC(4) receptor allowed some surface expression in the absence of an accessory protein, while ACTH-induced cAMP production remained entirely MRAP-dependent. On the other hand, replacing the last two transmembrane domains, third extracellular loop and C-terminal tail of the MC(4) receptor with the corresponding regions from the MC(2) receptor resulted in MRAP-dependent signaling. Surprisingly, replacing the second and third transmembrane domains and the intervening first extracellular loop of MC(2) receptors with MC(4) sequences generated a chimera (2C2) that responded to both adrenocorticotropic hormone (ACTH) and to the potent MSH analog 4-norleucine-7-d-phenylalanine-α-melanocyte stimulating hormone (NDP-α-MSH), which does not activate native MC(2) receptors. The 2C2 chimeric receptor was able to respond to NDP-α-MSH without MRAP, but MRAP shifted the EC50 value for NDP-α-MSH to the left and caused constitutive activity. These results identify the first transmembrane domain as important for surface expression and regions from the second to third transmembrane segments of the MC(2) receptor as important for MRAP dependent-signal transduction and ligand specificity.


Subject(s)
Membrane Proteins/metabolism , Receptor, Melanocortin, Type 2/chemistry , Receptor, Melanocortin, Type 2/metabolism , Receptor, Melanocortin, Type 4/chemistry , Receptor, Melanocortin, Type 4/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Humans , Ligands , Mice , Protein Transport , Receptor, Melanocortin, Type 2/genetics , Receptor, Melanocortin, Type 4/genetics , Recombinant Fusion Proteins/genetics , Signal Transduction , Substrate Specificity
19.
Sci Signal ; 3(116): ra28, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20371771

ABSTRACT

Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs), which constitute the largest family of membrane proteins, mediate responses to diverse physiological stimuli. The presence of melanocortin 2 receptors (MC2Rs) on the plasma membrane requires the presence of either MC2R accessory protein (MRAP) or MRAP2, which are homologous accessory proteins. Here, we show that, whereas MRAP was essential for activation of MC2R signaling, MRAP2 was an endogenous inhibitor that competed with MRAP for binding to MC2R and decreased the potency of adrenocorticotropic hormone (ACTH), the endogenous agonist for MC2Rs, in stimulating the production of adenosine 3',5'-monophosphate (cAMP). ACTH bound with high affinity to MC2Rs in the presence of MRAP, but not MRAP2. The ability of MRAP and MRAP2 to influence ligand-binding affinity was specific to MC2R, because these proteins had little effect on the binding of NDP-alpha-melanocyte-stimulating hormone to MC4R or on its stimulation of cAMP responses. These results demonstrate that the balance of stimulatory and inhibitory accessory proteins can control the sensitivity of a GPCR to its natural agonist.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Receptor, Melanocortin, Type 2/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Adrenocorticotropic Hormone/metabolism , Analysis of Variance , Animals , CHO Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cloning, Molecular , Cricetinae , Cricetulus , Cyclic AMP/biosynthesis , DNA Primers/genetics , Dimerization , Enzyme-Linked Immunosorbent Assay , Humans , Membrane Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction
20.
J Biol Chem ; 284(34): 22641-8, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19535343

ABSTRACT

MC2 (ACTH) receptors require MC2 receptor accessory protein (MRAP) to reach the cell surface. In this study, we show that MRAP has the opposite effect on the closely related MC5 receptor. In enzyme-linked immunosorbent assay and microscopy experiments, MC2 receptor was retained in the endoplasmic reticulum in the absence of MRAP and targeted to the plasma membrane with MRAP. MC5 receptor was at the plasma membrane in the absence of MRAP, but trapped intracellularly when expressed with MRAP. Using bimolecular fluorescence complementation, where one fragment of yellow fluorescent protein (YFP) was fused to receptors and another to MRAP, we showed that MC2 receptor-MRAP dimers were present at the plasma membrane, whereas MC5 receptor-MRAP dimers were intracellular. Both MC2 and MC5 receptors co-precipitated with MRAP. MRAP did not alter expression of beta2-adrenergic receptors or co-precipitate with them. To determine if MRAP affects formation of receptor oligomers, we co-expressed MC2 receptors fused to YFP fragments in the presence or absence of MRAP. YFP fluorescence, reporting MC2 receptor homodimers, was readily detectable with or without MRAP. In contrast, MC5 receptor homodimers were visible in the absence of MRAP, but little fluorescence was observed by microscopic analysis when MRAP was co-expressed. Co-precipitation of differentially tagged receptors confirmed that MRAP blocks MC5 receptor dimerization. The regions of MRAP required for its effects on MC2 and MC5 receptors differed. These results establish that MRAP forms stable complexes with two different melanocortin receptors, facilitating surface expression of MC2 receptor but disrupting dimerization and surface localization of MC5 receptor.


Subject(s)
Membrane Proteins/metabolism , Receptor, Melanocortin, Type 2/metabolism , Receptors, Melanocortin/metabolism , Animals , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Immunoprecipitation , Membrane Proteins/genetics , Mice , Protein Multimerization , Receptor, Melanocortin, Type 2/genetics , Receptors, Melanocortin/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...