Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cell Mol Life Sci ; 77(24): 5189-5205, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31900622

ABSTRACT

Transmission of extracellular signals by G protein-coupled receptors typically relies on a cascade of intracellular events initiated by the activation of heterotrimeric G proteins or ß-arrestins followed by effector activation/inhibition. Here, we report an alternative signal transduction mode used by the orphan GPR50 that relies on the nuclear translocation of its carboxyl-terminal domain (CTD). Activation of the calcium-dependent calpain protease cleaves off the CTD from the transmembrane-bound GPR50 core domain between Phe-408 and Ser-409 as determined by MALDI-TOF-mass spectrometry. The cytosolic CTD then translocates into the nucleus assisted by its 'DPD' motif, where it interacts with the general transcription factor TFII-I to regulate c-fos gene transcription. RNA-Seq analysis indicates a broad role of the CTD in modulating gene transcription with ~ 8000 differentially expressed genes. Our study describes a non-canonical, direct signaling mode of GPCRs to the nucleus with similarities to other receptor families such as the NOTCH receptor.


Subject(s)
Nerve Tissue Proteins/genetics , Protein Transport/genetics , Receptors, G-Protein-Coupled/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Humans , Protein Binding/genetics , Receptors, Notch , Signal Transduction/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
2.
J Pineal Res ; 66(2): e12540, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30475390

ABSTRACT

Melatonin receptors play important roles in the regulation of circadian and seasonal rhythms, sleep, retinal functions, the immune system, depression, and type 2 diabetes development. Melatonin receptors are approved drug targets for insomnia, non-24-hour sleep-wake disorders, and major depressive disorders. In mammals, two melatonin receptors (MTRs) exist, MT1 and MT2 , belonging to the G protein-coupled receptor (GPCR) superfamily. Similar to most other GPCRs, reliable antibodies recognizing melatonin receptors proved to be difficult to obtain. Here, we describe the development of the first monoclonal antibodies (mABs) for mouse MT1 and MT2 . Purified antibodies were extensively characterized for specific reactivity with mouse, rat, and human MT1 and MT2 by Western blot, immunoprecipitation, immunofluorescence, and proximity ligation assay. Several mABs were specific for either mouse MT1 or MT2 . None of the mABs cross-reacted with rat MTRs, and some were able to react with human MTRs. The specificity of the selected mABs was validated by immunofluorescence microscopy in three established locations (retina, suprachiasmatic nuclei, pituitary gland) for MTR expression in mice using MTR-KO mice as control. MT2 expression was not detected in mouse insulinoma MIN6 cells or pancreatic beta-cells. Collectively, we report the first monoclonal antibodies recognizing recombinant and native mouse melatonin receptors that will be valuable tools for future studies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Receptor, Melatonin, MT1/analysis , Receptor, Melatonin, MT2/analysis , Animals , Mice , Protein Domains , Receptor, Melatonin, MT1/immunology , Receptor, Melatonin, MT2/immunology
3.
J Biol Chem ; 290(18): 11537-46, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25770211

ABSTRACT

Inasmuch as the neurohormone melatonin is synthetically derived from serotonin (5-HT), a close interrelationship between both has long been suspected. The present study reveals a hitherto unrecognized cross-talk mediated via physical association of melatonin MT2 and 5-HT2C receptors into functional heteromers. This is of particular interest in light of the "synergistic" melatonin agonist/5-HT2C antagonist profile of the novel antidepressant agomelatine. A suite of co-immunoprecipitation, bioluminescence resonance energy transfer, and pharmacological techniques was exploited to demonstrate formation of functional MT2 and 5-HT2C receptor heteromers both in transfected cells and in human cortex and hippocampus. MT2/5-HT2C heteromers amplified the 5-HT-mediated Gq/phospholipase C response and triggered melatonin-induced unidirectional transactivation of the 5-HT2C protomer of MT2/5-HT2C heteromers. Pharmacological studies revealed distinct functional properties for agomelatine, which shows "biased signaling." These observations demonstrate the existence of functionally unique MT2/5-HT2C heteromers and suggest that the antidepressant agomelatine has a distinctive profile at these sites potentially involved in its therapeutic effects on major depression and generalized anxiety disorder. Finally, MT2/5-HT2C heteromers provide a new strategy for the discovery of novel agents for the treatment of psychiatric disorders.


Subject(s)
Melatonin/metabolism , Protein Multimerization , Receptor, Melatonin, MT2/chemistry , Receptor, Serotonin, 5-HT2C/chemistry , Serotonin/metabolism , Signal Transduction , Acetamides/pharmacology , Arrestins/metabolism , Drug Synergism , Gene Expression Regulation/drug effects , HEK293 Cells , HeLa Cells , Humans , Melatonin/pharmacology , Protein Multimerization/drug effects , Protein Structure, Quaternary , Protein Transport/drug effects , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin/pharmacology , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Type C Phospholipases/metabolism , beta-Arrestins
4.
Bioessays ; 36(8): 778-87, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24903552

ABSTRACT

Melatonin, the neuro-hormone synthesized during the night, has recently seen an unexpected extension of its functional implications toward type 2 diabetes development, visual functions, sleep disturbances, and depression. Transgenic mouse models were instrumental for the establishment of the link between melatonin and these major human diseases. Most of the actions of melatonin are mediated by two types of G protein-coupled receptors, named MT1 and MT2 , which are expressed in many different organs and tissues. Understanding the pharmacology and function of mouse MT1 and MT2 receptors, including MT1 /MT2 heteromers, will be of crucial importance to evaluate the relevance of these mouse models for future therapeutic developments. This review will critically discuss these aspects, and give some perspectives including the generation of new mouse models.


Subject(s)
Receptors, Melatonin/physiology , Animals , Circadian Rhythm , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Humans , Melatonin/physiology , Mice , Photoperiod , Signal Transduction , Sleep
5.
EMBO J ; 29(21): 3646-59, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-20859254

ABSTRACT

Functional asymmetry of G-protein-coupled receptor (GPCR) dimers has been reported for an increasing number of cases, but the molecular architecture of signalling units associated to these dimers remains unclear. Here, we characterized the molecular complex of the melatonin MT1 receptor, which directly and constitutively couples to G(i) proteins and the regulator of G-protein signalling (RGS) 20. The molecular organization of the ternary MT1/G(i)/RGS20 complex was monitored in its basal and activated state by bioluminescence resonance energy transfer between probes inserted at multiple sites of the complex. On the basis of the reported crystal structures of G(i) and the RGS domain, we propose a model wherein one G(i) and one RGS20 protein bind to separate protomers of MT1 dimers in a pre-associated complex that rearranges upon agonist activation. This model was further validated with MT1/MT2 heterodimers. Collectively, our data extend the concept of asymmetry within GPCR dimers, reinforce the notion of receptor specificity for RGS proteins and highlight the advantage of GPCRs organized as dimers in which each protomer fulfils its specific task by binding to different GPCR-interacting proteins.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTPase-Activating Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptor, Melatonin, MT1/metabolism , Amino Acid Sequence , Cells, Cultured , Electrophysiology , Energy Transfer , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Melatonin/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Protein Conformation , Protein Multimerization , RGS Proteins , Receptor, Melatonin, MT1/chemistry , Receptor, Melatonin, MT1/genetics , Signal Transduction
6.
Methods Mol Biol ; 2550: 179-188, 2022.
Article in English | MEDLINE | ID: mdl-36180691

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is an important ubiquitous second messenger and one of the major pathways transducing the activation of G protein-coupled receptors (GPCRs). Quantifying intracellular levels of cAMP in an accurate and high-throughput manner is, therefore, of high interest to access functional responses of GPCRs. The neurohormone melatonin is selectively recognized by two GPCRs in mammals, named MT1 and MT2. Both have an inhibitory action on intracellular cAMP levels. Here, we describe a homogeneous high-throughput-compatible methodology routinely used in our laboratory to measure cAMP levels following activation of melatonin receptors.


Subject(s)
Melatonin , Adenosine Monophosphate , Animals , Cyclic AMP/metabolism , Mammals/metabolism , Melatonin/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Melatonin
7.
ACS Pharmacol Transl Sci ; 5(2): 89-101, 2022 Feb 11.
Article in English | MEDLINE | ID: mdl-35846981

ABSTRACT

G protein-coupled receptors (GPCRs) can engage distinct subsets of signaling pathways, but the structural determinants of this functional selectivity remain elusive. The naturally occurring genetic variants of GPCRs, selectively affecting different pathways, offer an opportunity to explore this phenomenon. We previously identified 40 coding variants of the MTNR1B gene encoding the melatonin MT2 receptor (MT2). These mutations differently impact the ß-arrestin 2 recruitment, ERK activation, cAMP production, and Gαi1 and Gαz activation. In this study, we combined functional clustering and structural modeling to delineate the molecular features controlling the MT2 functional selectivity. Using non-negative matrix factorization, we analyzed the signaling signatures of the 40 MT2 variants yielding eight clusters defined by unique signaling features and localized in distinct domains of MT2. Using computational homology modeling, we describe how specific mutations can selectively affect the subsets of signaling pathways and offer a proof of principle that natural variants can be used to explore and understand the GPCR functional selectivity.

8.
J Pineal Res ; 51(4): 394-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21615493

ABSTRACT

Melatonin is a powerful antioxidant and a synchronizer of many physiological processes. Alteration in melatonin signaling has been reported in a broad range of diseases, but little is known about the genetic variability of this pathway in humans. Here, we sequenced all the genes of the melatonin pathway -AA-NAT, ASMT, MTNR1A, MTNR1B and GPR50 - in 321 individuals from Sweden including 101 patients with attention-deficit/hyperactivity disorder (ADHD) and 220 controls from the general population. We could find several damaging mutations in patients with ADHD, but no significant enrichment compared with the general population. Among these variations, we found a splice site mutation in ASMT (IVS5+2T>C) and one stop mutation in MTNR1A (Y170X) - detected exclusively in patients with ADHD - for which biochemical analyses indicated that they abolish the activity of ASMT and MTNR1A. These genetic and functional results represent the first comprehensive ascertainment of melatonin signaling deficiency in ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Genetic Variation/genetics , Melatonin/genetics , Acetylserotonin O-Methyltransferase/genetics , Arylalkylamine N-Acetyltransferase/genetics , Female , Humans , Male , Nerve Tissue Proteins/genetics , Receptor, Melatonin, MT1/genetics , Receptors, G-Protein-Coupled/genetics
9.
Mol Cell Proteomics ; 7(8): 1556-69, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18448421

ABSTRACT

G protein-coupled receptors (GPCRs) constitute the largest family of membrane receptors and are major drug targets. Recent progress has shown that GPCRs are part of large protein complexes that regulate their activity. We present here a generic approach for identification of these complexes that is based on the use of receptor subdomains and that overcomes the limitations of currently used genetics and proteomics approaches. Our approach consists of a carefully balanced combination of chemically synthesized His6-tagged baits, immobilized metal affinity chromatography, one- and two-dimensional gel electrophoresis separation and mass spectrometric identification. The carboxyl-terminal tails (C-tails) of the human MT1 and MT2 melatonin receptors, two class A GPCRs, were used as models to purify protein complexes from mouse brain lysates. We identified 32 proteins that interacted with the C-tail of MT1, 14 proteins that interacted with the C-tail of MT2, and eight proteins that interacted with both C-tails. Several randomly selected proteins were validated by Western blotting, and the functional relevance of our data was further confirmed by showing the interaction between the full-length MT1 and the regulator of G protein signaling Z1 in transfected HEK 293 cells and native tissue. Taken together, we have established an integrated and generic purification strategy for the identification of high quality and functionally relevant GPCR-associated protein complexes that significantly widens the repertoire of available techniques.


Subject(s)
Multiprotein Complexes/isolation & purification , Proteomics/methods , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Chromatography, Affinity , GTPase-Activating Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mice , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , RGS Proteins
10.
Br J Pharmacol ; 175(16): 3281-3297, 2018 08.
Article in English | MEDLINE | ID: mdl-28898928

ABSTRACT

BACKGROUND AND PURPOSE: Recent crystal structures of GPCRs have emphasized the previously unappreciated role of the second extracellular (E2) loop in ligand binding and gating and receptor activation. Here, we have assessed the role of the E2 loop in the activation of the melatonin MT1 receptor and in the inactivation of the closely related orphan receptor GPR50. EXPERIMENTAL APPROACH: Chimeric MT1 -GPR50 receptors were generated and functionally analysed in terms of 2-[125 I]iodomelatonin binding, Gi /cAMP signalling and ß-arrestin2 recruitment. We also used computational molecular dynamics (MD) simulations. KEY RESULTS: MD simulations of 300 ns revealed (i) the tight hairpin structure of the E2 loop of the MT1 receptor (ii) the most suitable features for melatonin binding in MT1 receptors and (iii) major predicted rearrangements upon MT1 receptor activation, stabilizing interaction networks between Phe179 or Gln181 in the E2 loop and transmembrane helixes 5 and 6. Functional assays confirmed these predictions, because reciprocal replacement of MT1 and GPR50 residues/domains led to the predicted loss- and gain-of-melatonin action of MT1 receptors and GPR50 respectively. CONCLUSIONS AND IMPLICATIONS: Our work demonstrated the crucial role of the E2 loop for MT1 receptor and GPR50 function by proposing a model in which the E2 loop is important in stabilizing active MT1 receptor conformations and by showing how evolutionary processes appear to have selected for modifications in the E2 loop in order to make GPR50 unresponsive to melatonin. LINKED ARTICLES: This article is part of a themed section on Recent Developments in Research of Melatonin and its Potential Therapeutic Applications. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.16/issuetoc.


Subject(s)
Receptor, Melatonin, MT1/chemistry , Receptor, Melatonin, MT1/metabolism , HEK293 Cells , Humans , Melatonin/metabolism , Models, Molecular , Nerve Tissue Proteins/metabolism , Protein Structure, Secondary , Receptors, G-Protein-Coupled/metabolism
11.
Sci Signal ; 11(545)2018 08 28.
Article in English | MEDLINE | ID: mdl-30154102

ABSTRACT

Melatonin is produced during the night and regulates sleep and circadian rhythms. Loss-of-function variants in MTNR1B, which encodes the melatonin receptor MT2, a G protein-coupled receptor (GPCR), are associated with an increased risk of type 2 diabetes (T2D). To identify specific T2D-associated signaling pathway(s), we profiled the signaling output of 40 MT2 variants by monitoring spontaneous (ligand-independent) and melatonin-induced activation of multiple signaling effectors. Genetic association analysis showed that defects in the melatonin-induced activation of Gαi1 and Gαz proteins and in spontaneous ß-arrestin2 recruitment to MT2 were the most statistically significantly associated with an increased T2D risk. Computational variant impact prediction by in silico evolutionary lineage analysis strongly correlated with the measured phenotypic effect of each variant, providing a predictive tool for future studies on GPCR variants. Together, this large-scale functional study provides an operational framework for the postgenomic analysis of the multiple GPCR variants present in the human population. The association of T2D risk with signaling pathway-specific defects opens avenues for pathway-specific personalized therapeutic intervention and reveals the potential relevance of MT2 function during the day, when melatonin is undetectable, but spontaneous activity of the receptor occurs.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Variation , Receptor, Melatonin, MT2/genetics , Signal Transduction/genetics , Antioxidants/pharmacology , Diabetes Mellitus, Type 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Melatonin/pharmacology , Phosphorylation/drug effects , Receptor, Melatonin, MT2/metabolism , Signal Transduction/drug effects , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
12.
Nat Commun ; 9(1): 1216, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572483

ABSTRACT

Transforming growth factor-ß (TGFß) signaling is initiated by the type I, II TGFß receptor (TßRI/TßRII) complex. Here we report the formation of an alternative complex between TßRI and the orphan GPR50, belonging to the G protein-coupled receptor super-family. The interaction of GPR50 with TßRI induces spontaneous TßRI-dependent Smad and non-Smad signaling by stabilizing the active TßRI conformation and competing for the binding of the negative regulator FKBP12 to TßRI. GPR50 overexpression in MDA-MB-231 cells mimics the anti-proliferative effect of TßRI and decreases tumor growth in a xenograft mouse model. Inversely, targeted deletion of GPR50 in the MMTV/Neu spontaneous mammary cancer model shows decreased survival after tumor onset and increased tumor growth. Low GPR50 expression is associated with poor survival prognosis in human breast cancer irrespective of the breast cancer subtype. This describes a previously unappreciated spontaneous TGFß-independent activation mode of TßRI and identifies GPR50 as a TßRI co-receptor with potential impact on cancer development.


Subject(s)
Mammary Neoplasms, Animal/prevention & control , Nerve Tissue Proteins/physiology , Receptor, Transforming Growth Factor-beta Type I/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Endosomes/metabolism , Female , Gene Expression Profiling , HEK293 Cells , HeLa Cells , Humans , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smad Proteins/metabolism , Tacrolimus Binding Protein 1A/metabolism
13.
Chronobiol Int ; 23(1-2): 419-26, 2006.
Article in English | MEDLINE | ID: mdl-16687315

ABSTRACT

In mammals, the circadian hormone melatonin targets two seven-transmembrane-spanning receptors, MT1 and MT2, of the G protein-coupled receptor (GPCR) super-family. Evidence accumulated over the last 15 yrs convincingly demonstrates that GPCRs, classically considered to function as monomers, are actually organized as homodimers and heterodimerize with other GPCR family members. These dimers are formed early in the biosynthetic pathway and remain stable throughout the entire life cycle. A growing number of observations demonstrate that GPCR oligomerization may occur in native tissues and may have important consequences on receptor function. The formation of MT1 and MT2 homodimers and MT1/MT2 heterodimers has been shown in heterologous expression systems at physiological expression levels. Formation of MT1/MT2 heterodimers remains to be shown in native tissues but is suggested by the documented co-expression of MT1 and MT2 in many melatonin-sensitive tissues, such as the hypothalamic suprachiasmatic nuclei, retina, arteries, and adipose tissue. Considering that multiple GPCRs are expressed simultaneously in most cells, the possible engagement into heterodimeric complexes has to be considered and taken into account for the interpretation of experimental data obtained from native tissues and knockout animals.


Subject(s)
Melatonin/chemistry , Receptors, G-Protein-Coupled/chemistry , Receptors, Melatonin/chemistry , Animals , Biological Clocks , Circadian Rhythm , Dimerization , Humans , Models, Biological , Models, Molecular , Receptor, Melatonin, MT1/physiology , Receptor, Melatonin, MT2/physiology
14.
Mol Endocrinol ; 27(8): 1217-33, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23798576

ABSTRACT

The existence of interindividual variations in G protein-coupled receptor sequences has been recognized early on. Recent advances in large-scale exon sequencing techniques are expected to dramatically increase the number of variants identified in G protein-coupled receptors, giving rise to new challenges regarding their functional characterization. The current minireview will illustrate these challenges based on the MTNR1B gene, which encodes the melatonin MT2 receptor, for which exon sequencing revealed 40 rare nonsynonymous variants in the general population and in type 2 diabetes (T2D) cohorts. Functional characterization of these MT2 mutants revealed 14 mutants with loss of Gi protein activation that associate with increased risk of T2D development. This repertoire of disease-associated mutants is a rich source for structure-activity studies and will help to define the still poorly understood role of melatonin in glucose homeostasis and T2D development in humans. Defining the functional defects in carriers of rare MT2 mutations will help to provide personalized therapies to these patients in the future.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Melatonin/metabolism , Receptor, Melatonin, MT2/metabolism , Amino Acid Sequence , Animals , Diabetes Mellitus, Type 2/genetics , Humans , Mice , Molecular Sequence Data , Obesity/metabolism , Pineal Gland/metabolism , Polymorphism, Single Nucleotide , Rats , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/genetics , Structure-Activity Relationship
15.
Sci Signal ; 6(296): ra89, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24106342

ABSTRACT

The formation of G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor (GPCR) heteromers enables signaling diversification and holds great promise for improved drug selectivity. Most studies of these oligomerization events have been conducted in heterologous expression systems, and in vivo validation is lacking in most cases, thus questioning the physiological significance of GPCR heteromerization. The melatonin receptors MT1 and MT2 exist as homomers and heteromers when expressed in cultured cells. We showed that melatonin MT1/MT2 heteromers mediated the effect of melatonin on the light sensitivity of rod photoreceptors in mice. This effect of melatonin involved activation of the heteromer-specific phospholipase C and protein kinase C (PLC/PKC) pathway and was abolished in MT1(-/-) or MT2(-/-) mice, as well as in mice overexpressing a nonfunctional MT2 mutant that interfered with the formation of functional MT1/MT2 heteromers in photoreceptor cells. Not only does this study establish an essential role of melatonin receptor heteromers in retinal function, it also provides in vivo support for the physiological importance of GPCR heteromerization. Thus, the MT1/MT2 heteromer complex may provide a specific pharmacological target to improve photoreceptor function.


Subject(s)
Eye Proteins/metabolism , Protein Multimerization/physiology , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Eye Proteins/genetics , Mice , Mice, Knockout , Mutation , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics , Retinal Rod Photoreceptor Cells/cytology , Type C Phospholipases/genetics , Type C Phospholipases/metabolism
16.
Nat Genet ; 44(3): 297-301, 2012 Jan 29.
Article in English | MEDLINE | ID: mdl-22286214

ABSTRACT

Genome-wide association studies have revealed that common noncoding variants in MTNR1B (encoding melatonin receptor 1B, also known as MT(2)) increase type 2 diabetes (T2D) risk(1,2). Although the strongest association signal was highly significant (P < 1 × 10(-20)), its contribution to T2D risk was modest (odds ratio (OR) of ∼1.10-1.15)(1-3). We performed large-scale exon resequencing in 7,632 Europeans, including 2,186 individuals with T2D, and identified 40 nonsynonymous variants, including 36 very rare variants (minor allele frequency (MAF) <0.1%), associated with T2D (OR = 3.31, 95% confidence interval (CI) = 1.78-6.18; P = 1.64 × 10(-4)). A four-tiered functional investigation of all 40 mutants revealed that 14 were non-functional and rare (MAF < 1%), and 4 were very rare with complete loss of melatonin binding and signaling capabilities. Among the very rare variants, the partial- or total-loss-of-function variants but not the neutral ones contributed to T2D (OR = 5.67, CI = 2.17-14.82; P = 4.09 × 10(-4)). Genotyping the four complete loss-of-function variants in 11,854 additional individuals revealed their association with T2D risk (8,153 individuals with T2D and 10,100 controls; OR = 3.88, CI = 1.49-10.07; P = 5.37 × 10(-3)). This study establishes a firm functional link between MTNR1B and T2D risk.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Variation , Receptor, Melatonin, MT2/genetics , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Body Mass Index , Diabetes Mellitus, Type 2/metabolism , Exons/genetics , Gene Frequency , Genome-Wide Association Study , Humans , Logistic Models , Models, Molecular , Molecular Sequence Data , Odds Ratio , Sequence Analysis, DNA , White People/genetics
17.
Adv Pharmacol ; 62: 349-80, 2011.
Article in English | MEDLINE | ID: mdl-21907915

ABSTRACT

G protein-coupled receptors (GPCRs) are, with approximately 800 members, among the most abundant membrane proteins in humans. They are responding to a plethora of ligands and are involved in the transmission of extracellular signals inside the cell. GPCRs are synthesized in the endoplasmatic reticulum and are then transported to the cell surface where they are typically activated. Receptor activation triggers several processes such as signaling and receptor endocytosis. Along their life cycle, GPCRs are accompanied by a range of specialized GPCR-interacting proteins (GIPs) to assist nascent receptors in proper folding, to target them to the appropriate subcellular compartments and to fulfill their signaling tasks. Differential expression of GIPs and rapid alterations of GPCR/GIP interaction networks are efficient means to regulate GPCR function in a tissue-specific and spatiotemporal manner to trigger appropriate cellular responses. Interfering with a GPCR/GIP interaction might become a new strategy for specific therapeutic intervention. This chapter will focus on the importance of GIPs along the GPCR life cycle and discuss the dynamics and molecular organization of GPCR/GIP complexes.


Subject(s)
Carrier Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Membrane/metabolism , Endocytosis , Humans , Protein Binding , Receptors, G-Protein-Coupled/biosynthesis , Signal Transduction
18.
PLoS One ; 5(7): e11495, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20657642

ABSTRACT

Melatonin is a powerful antioxidant and a synchronizer of many physiological processes. Alteration of the melatonin pathway has been reported in circadian disorders, diabetes and autism spectrum disorders (ASD). However, very little is known about the genetic variability of melatonin receptors in humans. Here, we sequenced the melatonin receptor MTNR1A and MTNR1B, genes coding for MT1 and MT2 receptors, respectively, in a large panel of 941 individuals including 295 patients with ASD, 362 controls and 284 individuals from different ethnic backgrounds. We also sequenced GPR50, coding for the orphan melatonin-related receptor GPR50 in patients and controls. We identified six non-synonymous mutations for MTNR1A and ten for MTNR1B. The majority of these variations altered receptor function. Particularly interesting mutants are MT1-I49N, which is devoid of any melatonin binding and cell surface expression, and MT1-G166E and MT1-I212T, which showed severely impaired cell surface expression. Of note, several mutants possessed pathway-selective signaling properties, some preferentially inhibiting the adenylyl cyclase pathway, others preferentially activating the MAPK pathway. The prevalence of these deleterious mutations in cases and controls indicates that they do not represent major risk factor for ASD (MTNR1A case 3.6% vs controls 4.4%; MTNR1B case 4.7% vs 3% controls). Concerning GPR50, we detected a significant association between ASD and two variations, Delta502-505 and T532A, in affected males, but it did not hold up after Bonferonni correction for multiple testing. Our results represent the first functional ascertainment of melatonin receptors in humans and constitute a basis for future structure-function studies and for interpreting genetic data on the melatonin pathway in patients.


Subject(s)
Child Development Disorders, Pervasive/genetics , Receptors, Melatonin/genetics , Adult , Animals , COS Cells , Cell Line , Child , Chlorocebus aethiops , Cyclic AMP/metabolism , Female , Humans , Male , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation/genetics , Receptor, Melatonin, MT1/genetics , Receptor, Melatonin, MT2/genetics
19.
J Biol Chem ; 283(24): 16762-71, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18378672

ABSTRACT

Intracellular signaling events are often organized around PDZ (PSD-95/Drosophila Disc large/ZO-1 homology) domain-containing scaffolding proteins. The ubiquitously expressed multi-PDZ protein MUPP1, which is composed of 13 PDZ domains, has been shown to interact with multiple viral and cellular proteins and to play important roles in receptor targeting and trafficking. In this study, we show that MUPP1 binds to the G protein-coupled MT(1) melatonin receptor and directly regulates its G(i)-dependent signal transduction. Structural determinants involved in this interaction are the PDZ10 domain of MUPP1 and the valine of the canonical class III PDZ domain binding motif DSV of the MT(1) carboxyl terminus. This high affinity interaction (K(d) approximately 4 nm), which is independent of MT(1) activation, occurs in the ovine pars tuberalis of the pituitary expressing both proteins endogenously. Although the disruption of the MT(1)/MUPP1 interaction has no effect on the subcellular localization, trafficking, or degradation of MT(1), it destabilizes the interaction between MT(1) and G(i) and abolishes G(i)-mediated signaling of MT(1). Our findings highlight a previously unappreciated role of PDZ proteins in promoting G protein coupling to receptors.


Subject(s)
Carrier Proteins/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptor, Melatonin, MT1/metabolism , Amino Acid Motifs , Animals , Carrier Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Kinetics , Models, Biological , Pituitary Gland/metabolism , Protein Binding , Receptors, Melatonin/metabolism , Sheep , Signal Transduction , Two-Hybrid System Techniques , Valine/chemistry
20.
Mol Cell Proteomics ; 6(5): 835-44, 2007 May.
Article in English | MEDLINE | ID: mdl-17215244

ABSTRACT

G protein-coupled receptors (GPCRs) constitute the largest family of membrane receptors and are of major therapeutic importance. The identification of GPCR-associated proteins is an important step toward a better understanding of these receptors. However, current methods are not satisfying as only isolated receptor domains (intracellular loops or carboxyl-terminal tails) can be used as "bait." We report here a method based on tandem affinity purification coupled to mass spectrometry that overcomes these limitations as the entire receptor is used to identify protein complexes formed in living mammalian cells. The human MT(1) and MT(2) melatonin receptors were chosen as model GPCRs. Both receptors were tagged with the tandem affinity purification tag at their carboxyl-terminal tails and expressed in human embryonic kidney 293 cells. Receptor solubilization and purification conditions were optimized. The method was validated by the co-purification of G(i) proteins, which are well known GPCR interaction partners but which are difficult to identify with current protein-protein interaction assays. Several new and functionally relevant MT(1)- and MT(2)-associated proteins were identified; some of them were common to both receptors, and others were specific for each subtype. Taken together, our protocol allowed for the first time the purification of GPCR-associated proteins under native conditions in quantities suitable for mass spectrometry analysis.


Subject(s)
Multiprotein Complexes/isolation & purification , Proteins/isolation & purification , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/metabolism , Cell Line , Chromatography, Affinity , Chromatography, Liquid , Humans , Mass Spectrometry , Receptor, Melatonin, MT1/chemistry , Receptor, Melatonin, MT2/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL