Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Commun Biol ; 4(1): 1070, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34522000

ABSTRACT

G protein-coupled receptors (GPCRs) are notoriously difficult to detect in native tissues. In an effort to resolve this problem, we have developed a novel mouse model by fusing the hemagglutinin (HA)-epitope tag sequence to the amino-terminus of the µ-opioid receptor (MOP). Although HA-MOP knock-in mice exhibit reduced receptor expression, we found that this approach allowed for highly efficient immunodetection of low abundant GPCR targets. We also show that the HA-tag facilitates both high-resolution imaging and immunoisolation of MOP. Mass spectrometry (MS) confirmed post-translational modifications, most notably agonist-selective phosphorylation of carboxyl-terminal serine and threonine residues. MS also unequivocally identified the carboxyl-terminal 387LENLEAETAPLP398 motif, which is part of the canonical MOP sequence. Unexpectedly, MS analysis of brain lysates failed to detect any of the 15 MOP isoforms that have been proposed to arise from alternative splicing of the MOP carboxyl-terminus. For quantitative analysis, we performed multiple successive rounds of immunodepletion using the well-characterized rabbit monoclonal antibody UMB-3 that selectively detects the 387LENLEAETAPLP398 motif. We found that >98% of HA-tagged MOP contain the UMB-3 epitope indicating that virtually all MOP expressed in the mouse brain exhibit the canonical amino acid sequence.


Subject(s)
Hemagglutinins/genetics , Receptors, Opioid, mu/genetics , Amino Acid Sequence , Animals , Female , Hemagglutinins/metabolism , Male , Mice , Phosphorylation , Protein Isoforms , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism
2.
Genome Biol Evol ; 13(8)2021 08 03.
Article in English | MEDLINE | ID: mdl-34137817

ABSTRACT

The large spectrum of hearing sensitivity observed in primates results from the impact of environmental and behavioral pressures to optimize sound perception and localization. Although evidence of positive selection in auditory genes has been detected in mammals including in Hominoids, selection has never been investigated in other primates. We analyzed 123 genes highly expressed in the inner ear of 27 primate species and tested to what extent positive selection may have shaped these genes in the order Primates tree. We combined both site and branch-site tests to obtain a comprehensive picture of the positively selected genes (PSGs) involved in hearing sensitivity, and drew a detailed description of the most affected branches in the tree. We chose a conservative approach, and thus focused on confounding factors potentially affecting PSG signals (alignment, GC-biased gene conversion, duplications, heterogeneous sequencing qualities). Using site tests, we showed that around 12% of these genes are PSGs, an α selection value consistent with average human genome estimates (10-15%). Using branch-site tests, we showed that the primate tree is heterogeneously affected by positive selection, with the black snub-nosed monkey, the bushbaby, and the orangutan, being the most impacted branches. A large proportion of these genes is inclined to shape hair cells and stereocilia, which are involved in the mechanotransduction process, known to influence frequency perception. Adaptive selection, and more specifically recurrent adaptive evolution, could have acted in parallel on a set of genes (ADGRV1, USH2A, PCDH15, PTPRQ, and ATP8A2) involved in stereocilia growth and the whole complex of bundle links connecting them, in species across different habitats, including high altitude and nocturnal environments.


Subject(s)
Mechanotransduction, Cellular , Stereocilia , Animals , Hair Cells, Auditory/physiology , Hearing/genetics , Primates/genetics
3.
J Proteomics ; 231: 104045, 2021 01 16.
Article in English | MEDLINE | ID: mdl-33189847

ABSTRACT

In the context of human evolution, the study of proteins may overcome the limitation of the high degradation of ancient DNA over time to provide biomolecular information useful for the phylogenetic reconstruction of hominid taxa. In this study, we used a shotgun proteomics approach to compare the tooth proteomes of extant human and non-human primates (gorilla, chimpanzee, orangutan and baboon) in order to search for a panel of peptides able to discriminate between taxa and further help reconstructing the evolutionary relationships of fossil primates. Among the 25 proteins shared by the five genera datasets, we found a combination of peptides with sequence variations allowing to differentiate the hominid taxa in the proteins AHSG, AMBN, APOA1, BGN, C9, COL11A2, COL22A1, COL3A1, DSPP, F2, LUM, OMD, PCOLCE and SERPINA1. The phylogenetic tree confirms the placement of the samples in the appropriate genus branches. Altogether, the results provide experimental evidence that a shotgun proteomics approach on dental tissue has the potential to detect taxonomic variation, which is promising for future investigations of uncharacterized and/or fossil hominid/hominin specimens. SIGNIFICANCE: A shotgun proteomics approach on human and non-human primate teeth allowed to identify peptides with taxonomic interest, highlighting the potential for future studies on hominid fossils.


Subject(s)
Hominidae , Tooth , Animals , Humans , Phylogeny , Primates , Proteome
4.
Peptides ; 134: 170406, 2020 12.
Article in English | MEDLINE | ID: mdl-32920044

ABSTRACT

The neuropeptide FF2 (NPFF2) receptor, predominantly expressed in the central nervous system, plays an important role in the modulation of sensory input and opioid analgesia, as well as in locomotion, feeding, intestinal motility, reward, and the control of obesity. The NPFF2 receptor belongs to the RFamide peptide receptor family and to the G protein coupled receptor (GPCR) super family, but contrary to many other class A GPCRs, no 3D structure has been solved. Thus, it is essential to perform mutagenesis to gain information on the fine functioning of the NPFF2 receptor. In this study, we examined the role of aspartic acid (D) from the "D/ERY/F" motif found in the second intracellular loop (ICL2) and the role of the C-terminal end of the receptor in ligand binding and signal transduction. We found that mutation D3.49A does not impair binding capacities but inhibits G protein activation as well as adenylyl cyclase regulation. Truncation of the C terminal part of the receptor has different effects depending on the position of truncation. When truncation was realized downstream of the putative acylation site, ligand binding and signal transduction capabilities were not lost, contrary to total deletion of the C terminus, which totally impairs the activity of the receptor.


Subject(s)
Analgesics, Opioid/pharmacology , Neuropeptides/pharmacology , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Cricetinae , Humans , Mutagenesis , Receptors, Neuropeptide/genetics , Signal Transduction/drug effects , Structure-Activity Relationship
5.
J Proteomics ; 211: 103548, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31626997

ABSTRACT

The study demonstrates the high potential of MS-based proteomics coupled to an iterative database search strategy for the in-depth investigation of ancient proteomes. An efficient targeted PRM MS-based approach, although limited to the detection of a single pair of sex-specific amelogenin peptides, allowed confirming the sex of individuals in ancient dental remains, an essential information for paleoanthropologists facing the issue of sex determination and dimorphism.


Subject(s)
Proteomics , Tooth , Amelogenin/genetics , Female , Humans , Male , Peptides , Sex Determination Analysis
6.
Peptides ; 122: 170154, 2019 12.
Article in English | MEDLINE | ID: mdl-31560950

ABSTRACT

Given the importance of G-protein coupled receptors in the regulation of many physiological functions, deciphering the relationships between genotype and phenotype in past and present hominin GPCRs is of main interest to understand the evolutionary process that contributed to the present-day variability in human traits and health. Here, we carefully examined the publicly available genomic and protein sequence databases of the archaic hominins (Neanderthal and Denisova) to draw up the catalog of coding variations in GPCRs for peptide ligands, in comparison with living humans. We then searched in the literature the functional changes, phenotypes and risk of disease possibly associated with the detected variants. Our survey suggests that Neanderthal and Denisovan hominins were likely prone to lower risk of obesity, to enhanced platelet aggregation in response to thrombin, to better response to infection, to less anxiety and aggressiveness and to favorable sociability. While some archaic variants were likely advantageous in the past, they might be responsible for maladaptive disorders today in the context of modern life and/or specific regional distribution. For example, an archaic haplotype in the neuromedin receptor 2 is susceptible to confer risk of diabetic nephropathy in type 1 diabetes in present-day Europeans. Paying attention to the pharmacological properties of some of the archaic variants described in this study may be helpful to understand the variability of therapeutic efficacy between individuals or ethnic groups.


Subject(s)
Diabetic Nephropathies/genetics , Evolution, Molecular , Obesity/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Diabetic Nephropathies/pathology , Genome, Human/genetics , Haplotypes/genetics , Hominidae/genetics , Humans , Neanderthals/genetics , Obesity/pathology , Peptides/genetics , Platelet Aggregation/genetics , Risk Factors
7.
PLoS One ; 12(9): e0183802, 2017.
Article in English | MEDLINE | ID: mdl-28902892

ABSTRACT

Environment parameters, diet and genetic factors interact to shape tooth morphostructure. In the human lineage, archaic and modern hominins show differences in dental traits, including enamel thickness, but variability also exists among living populations. Several polymorphisms, in particular in the non-collagenous extracellular matrix proteins of the tooth hard tissues, like enamelin, are involved in dental structure variation and defects and may be associated with dental disorders or susceptibility to caries. To gain insights into the relationships between tooth protein polymorphisms and dental structural morphology and defects, we searched for non-synonymous polymorphisms in tooth proteins from Neanderthal and Denisova hominins. The objective was to identify archaic-specific missense variants that may explain the dental morphostructural variability between extinct and modern humans, and to explore their putative impact on present-day dental phenotypes. Thirteen non-collagenous extracellular matrix proteins specific to hard dental tissues have been selected, searched in the publicly available sequence databases of Neanderthal and Denisova individuals and compared with modern human genome data. A total of 16 non-synonymous polymorphisms were identified in 6 proteins (ameloblastin, amelotin, cementum protein 1, dentin matrix acidic phosphoprotein 1, enamelin and matrix Gla protein). Most of them are encoded by dentin and enamel genes located on chromosome 4, previously reported to show signs of archaic introgression within Africa. Among the variants shared with modern humans, two are ancestral (common with apes) and one is the derived enamelin major variant, T648I (rs7671281), associated with a thinner enamel and specific to the Homo lineage. All the others are specific to Neanderthals and Denisova, and are found at a very low frequency in modern Africans or East and South Asians, suggesting that they may be related to particular dental traits or disease susceptibility in these populations. This modern regional distribution of archaic dental polymorphisms may reflect persistence of archaic variants in some populations and may contribute in part to the geographic dental variations described in modern humans.


Subject(s)
Dental Enamel Proteins/genetics , Hominidae , Neanderthals , Polymorphism, Genetic , Tooth/metabolism , Animals , Dental Enamel/anatomy & histology , Dental Enamel/metabolism , Dental Enamel Proteins/metabolism , Fossils , Gene Frequency , Genome, Human , Geography , Hominidae/genetics , Hominidae/metabolism , Humans , Neanderthals/genetics , Neanderthals/metabolism , Organ Size , Phylogeny , Selection, Genetic , Sequence Homology, Amino Acid , Tooth/anatomy & histology , Tooth/chemistry
8.
J Med Chem ; 59(5): 1925-45, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26824643

ABSTRACT

Derivatization of biologically active peptides by conjugation with fluorophores or radionuclide-bearing moieties is an effective and commonly used approach to prepare molecular tools and diagnostic agents. Whereas lysine, cysteine, and N-terminal amino acids have been mostly used for peptide conjugation, we describe a new, widely applicable approach to peptide conjugation based on the nonclassical bioisosteric replacement of the guanidine group in arginine by a functionalized carbamoylguanidine moiety. Four arginine-containing peptide receptor ligands (angiotensin II, neurotensin(8-13), an analogue of the C-terminal pentapeptide of neuropeptide Y, and a neuropeptide FF analogue) were subject of this proof-of-concept study. The N(ω)-carbamoylated arginines, bearing spacers with a terminal amino group, were incorporated into the peptides by standard Fmoc solid phase peptide synthesis. The synthesized chemically stable peptide derivatives showed high receptor affinities with Ki values in the low nanomolar range, even when bulky fluorophores had been attached. Two new tritiated tracers for angiotensin and neurotensin receptors are described.


Subject(s)
Angiotensin II/pharmacology , Arginine/analogs & derivatives , Neuropeptide Y/pharmacology , Neurotensin/pharmacology , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Receptors, Angiotensin/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Neurotensin/metabolism , Angiotensin II/chemistry , Arginine/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Structure , Neuropeptide Y/chemistry , Neurotensin/chemistry , Oligopeptides/chemistry , Peptide Fragments/chemistry , Receptors, Neuropeptide/agonists , Receptors, Neurotensin/antagonists & inhibitors , Structure-Activity Relationship
9.
Chem Biol Drug Des ; 87(6): 885-94, 2016 06.
Article in English | MEDLINE | ID: mdl-26802437

ABSTRACT

Methionine-enkephalin-Arg-Phe is an endogenous amphiactive analgesic peptide. Neuropeptide FF, on the other hand, is reported for its role in opioid modulation and tolerance development. Based on these reports, in the present study we designed a chimeric peptide NPYFa (YGGFMKKKPQRFamide), having the Met-enkephalin (opioid) and PQRFamide sequence of neuropeptide FF, which can then target both the opioid and neuropeptide FF receptors. We hypothesized that the chimeric peptide so designed would have both analgesic properties and further aid in understanding of the role of neuropeptide FF in the development of opiate tolerance. Our studies indicated that NPYFa induced an early onset, potent, dose-dependent and prolonged antinociception. Additionally, antagonists (MOR, KOR, and DOR) pretreatment studies determined a KOR-mediated antinociception activity of the ligand. Further, in vitro binding studies using the Eu-GTP-γS binding assay on cell lines expressing opioid and NPFF receptors showed binding to both the opioid and neuropeptide FF receptors suggesting a multiple receptor binding character of NPYFa. Moreover, chronic (6 days) treatment with NPYFa exhibited an absence of tolerance development subsequent to its analgesia. The current study proposes NPYFa as a potent, long-acting antinociceptor lacking tolerance development as well as a probe to study opioid analgesia and the associated complex mechanisms of tolerance development.


Subject(s)
Analgesia , Analgesics , Enkephalin, Methionine , Oligopeptides , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Dose-Response Relationship, Drug , Enkephalin, Methionine/analogs & derivatives , Enkephalin, Methionine/chemistry , Enkephalin, Methionine/pharmacokinetics , Enkephalin, Methionine/pharmacology , Male , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Rats , Rats, Wistar , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Time Factors
10.
J Med Chem ; 58(22): 8834-49, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26466164

ABSTRACT

Analogues of the argininamide-type NPY Y1 receptor (Y1R) antagonist BIBP3226, bearing carbamoyl moieties at the guanidine group, revealed subnanomolar Ki values and caused depression of the maximal response to NPY (calcium assay) by up to 90% in a concentration- and time-dependent manner, suggesting insurmountable antagonism. To gain insight into the mechanism of binding of the synthesized compounds, a tritiated antagonist, (R)-N(α)-diphenylacetyl-N(ω)-[2-([2,3-(3)H]propionylamino)ethyl]aminocarbonyl-(4-hydroxybenzyl)arginin-amide ([(3)H]UR-MK299, [(3)H]38), was prepared. [(3)H]38 revealed a dissociation constant in the picomolar range (Kd 0.044 nM, SK-N-MC cells) and very high Y1R selectivity. Apart from superior affinity, a considerably lower target off-rate (t1/2 95 min) was characteristic of [(3)H]38 compared to that of the higher homologue containing a tetramethylene instead of an ethylene spacer (t1/2 3 min, Kd 2.0 nM). Y1R binding of [(3)H]38 was fully reversible and fully displaceable by nonpeptide antagonists and the agonist pNPY. Therefore, the insurmountable antagonism observed in the functional assay has to be attributed to the extended target-residence time, a phenomenon of relevance in drug research beyond the NPY receptor field.


Subject(s)
Arginine/analogs & derivatives , Arginine/chemistry , Diphenylacetic Acids/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Receptors, Neuropeptide Y/antagonists & inhibitors , Amides , Animals , Arginine/pharmacokinetics , Binding, Competitive , CHO Cells , Cell Line , Cricetinae , Cricetulus , Fluorescent Dyes , Fura-2 , Half-Life , Humans , Isotope Labeling , Molecular Probes , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/pharmacology , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide Y/administration & dosage , Structure-Activity Relationship
11.
FEBS Lett ; 589(18): 2401-8, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26226422

ABSTRACT

Many in vitro data have shown that the efficacy of several opioid drugs is correlated with differential mu-opioid (MOP) receptor phosphorylation. Label-free semiquantitative on-line nanoflow liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) analyses were performed to compare the endogenous MOP receptor phosphorylation patterns of mice administered with morphine, etonitazene and fentanyl. The analysis identified S363, T370 and S375 as phosphorylated residues in the carboxy-terminus. Only T370 and S375 were regulated by agonists, with a higher propensity to promote double phosphorylation for high efficacy agonists. Our study provides confirmation that differential agonist-driven multi-site phosphorylation of MOP receptor occurs in vivo and validate the use of MS to study endogenous GPCR phosphorylation.


Subject(s)
Brain/metabolism , Phosphoproteins/metabolism , Proteomics , Receptors, Opioid, mu/metabolism , Amino Acid Sequence , Analgesics, Opioid/pharmacology , Animals , Brain/drug effects , Mass Spectrometry , Mice , Molecular Sequence Data , Phosphoproteins/agonists , Phosphoproteins/chemistry , Phosphorylation/drug effects , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry
12.
ACS Chem Neurosci ; 6(3): 438-45, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25588572

ABSTRACT

Through the development of a new class of unnatural ornithine derivatives as bioisosteres of arginine, we have designed an orally active peptidomimetic antagonist of neuropeptide FF receptors (NPFFR). Systemic low-dose administration of this compound to rats blocked opioid-induced hyperalgesia, without any apparent side-effects. Interestingly, we also observed that this compound potentiated opioid-induced analgesia. This unnatural ornithine derivative provides a novel therapeutic approach for both improving analgesia and reducing hyperalgesia induced by opioids in patients being treated for chronic pain.


Subject(s)
Analgesics, Opioid/toxicity , Fentanyl/toxicity , Hyperalgesia/chemically induced , Hyperalgesia/prevention & control , Peptidomimetics/therapeutic use , Animals , Arginine/metabolism , Chemical Phenomena , Cyclic AMP/metabolism , HEK293 Cells , Humans , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Ornithine/metabolism , Pain Threshold/drug effects , Peptidomimetics/chemistry , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/metabolism , Structure-Activity Relationship , Time Factors , Tritium/pharmacokinetics
13.
J Biol Chem ; 289(49): 33754-66, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25326382

ABSTRACT

The neuropeptide FF2 (NPFF2) receptor belongs to the rhodopsin family of G protein-coupled receptors and mediates the effects of several related RFamide neuropeptides. One of the main pharmacological interests of this system resides in its ability to regulate endogenous opioid systems, making it a potential target to reduce the negative effects of chronic opioid use. Phosphorylation of intracellular residues is the most extensively studied post-translational modification regulating G protein-coupled receptor activity. However, until now, no information concerning NPFF2 receptor phosphorylation is available. In this study, we combined mass spectrometric analysis and site-directed mutagenesis to analyze for the first time the phosphorylation pattern of the NPFF2 receptor and the role of the various phosphorylation sites in receptor signaling, desensitization, and trafficking in a SH-SY5Y model cell line. We identified the major, likely GRK-dependent, phosphorylation cluster responsible for acute desensitization, (412)TNST(415) at the end of the C terminus of the receptor, and additional sites involved in desensitization ((372)TS(373)) and internalization (Ser(395)). We thus demonstrate the key role played by phosphorylation in the regulation of NPFF2 receptor activity and trafficking. Our data also provide additional evidence supporting the concept that desensitization and internalization are partially independent processes relying on distinct phosphorylation patterns.


Subject(s)
Neurons/metabolism , Oligopeptides/metabolism , Protein Processing, Post-Translational , Receptors, Neuropeptide/chemistry , Amino Acid Sequence , Cell Line, Tumor , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Neurons/cytology , Oligopeptides/chemistry , Peptide Mapping , Phosphorylation , Protein Transport , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Sequence Alignment , Signal Transduction
14.
J Med Chem ; 57(21): 8903-27, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25268943

ABSTRACT

Neuropeptide FF1 and FF2 receptors (NPFF1-R and NPFF2-R), and their endogenous ligand NPFF, are one of only several systems responsible for mediating opioid-induced hyperalgesia, tolerance, and dependence. Currently, no small molecules displaying good affinity or selectivity for either subtype have been reported, to decipher the role of NPFF2-R as it relates to opioid-mediated analgesia, for further exploration of NPFF1-R, or for medication development for either subtype. We report the first nonpeptide small molecule scaffold for NPFF1,2-R, the guanidino-piperidines, and SAR studies resulting in the discovery of a NPFF1 agonist (7b, K(i) = 487 ± 117 nM), a NPFF1 antagonist (46, K(i) = 81 ± 17 nM), and a NPFF2 partial antagonist (53a, K(i) = 30 ± 5 nM), which serve as leads for the development of pharmacological probes and potential therapeutic agents. Testing of 46 alone was without effect in the mouse 48 °C warm-water tail-withdrawal test, but pretreatment with 46 prevented NPFF-induced hyperalgesia.


Subject(s)
Guanidines/chemical synthesis , Naphthalenes/chemical synthesis , Piperidines/chemical synthesis , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/antagonists & inhibitors , Animals , CHO Cells , Cricetulus , Guanidines/metabolism , HEK293 Cells , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Naphthalenes/metabolism , Piperidines/metabolism , Rats , Receptors, Opioid/metabolism , Structure-Activity Relationship
15.
J Biol Chem ; 289(41): 28697-706, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25183007

ABSTRACT

The dynamic organization of G protein-coupled receptors in the plasma membrane is suspected of playing a role in their function. The regulation of the diffusion mode of the mu-opioid (MOP) receptor was previously shown to be agonist-specific. Here we investigate the regulation of MOP receptor diffusion by heterologous activation of other G protein-coupled receptors and characterize the dynamic properties of the MOP receptor within the heterodimer MOP/neuropeptide FF (NPFF2) receptor. The data show that the dynamics and signaling of the MOP receptor in SH-SY5Y cells are modified by the activation of α2-adrenergic and NPFF2 receptors, but not by the activation of receptors not described to interact with the opioid receptor. By combining, for the first time, fluorescence recovery after photobleaching at variable radius experiments with bimolecular fluorescence complementation, we show that the MOP/NPFF2 heterodimer adopts a specific diffusion behavior that corresponds to a mix of the dynamic properties of both MOP and NPFF2 receptors. Altogether, the data suggest that heterologous regulation is accompanied by a specific organization of receptors in the membrane.


Subject(s)
Analgesics, Opioid/pharmacology , Protein Transport/drug effects , Receptor Cross-Talk/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Opioid, mu/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Clonidine/pharmacology , Diffusion , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Fluorescence Recovery After Photobleaching , Fluorescent Dyes , Gene Expression Regulation , Humans , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/pharmacology , Oligopeptides/pharmacology , Protein Multimerization , Receptors, Adrenergic, alpha-2/genetics , Receptors, Neuropeptide/genetics , Receptors, Opioid, mu/genetics , Signal Transduction
16.
Peptides ; 55: 79-84, 2014 May.
Article in English | MEDLINE | ID: mdl-24582609

ABSTRACT

The zwitterionic detergent CHAPS was used to solubilize the human mu-opioid receptor (hMOR) from SH-SY5Y neuroblastoma cells and recombinant hMOR-CHO (CHO-T7-hMOR) and hMOR-SH-SY5Y (SH-SY5Y-T7-hMOR) cell membranes. Agonist stimulation and G-protein activation by the mu-selective opioid agonist DAMGO ([D-Ala2, N-MePhe4, Gly-ol]-enkephalin) were recovered after removing of CHAPS after polyethylene glycol (PEG) precipitation. Binding assays show that hMOR solubilized and reconstituted this way was functional and able to interact with both agonist peptides and with G-protein. The effective solubilization and reconstitution of hMOR from mammalian cells, without truncation and extensive modification, represent an essential step toward the purification of a receptor bearing important post-translational modifications.


Subject(s)
Receptors, Opioid, mu/chemistry , Animals , CHO Cells , Cell Line, Tumor , Cholic Acids/chemistry , Cricetulus , Detergents/chemistry , Diprenorphine/chemistry , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , Humans , Ligands , Narcotic Antagonists/chemistry , Polyethylene Glycols/chemistry , Protein Binding , Protein Refolding , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/biosynthesis , Solubility
17.
Biol Psychiatry ; 76(10): 767-74, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-24629717

ABSTRACT

BACKGROUND: The clinical benefits of opioid drugs are counteracted by the development of tolerance and addiction. We provide in vivo evidence for the involvement of G protein-coupled receptor kinases (GRKs) in opioid dependence in addition to their roles in agonist-selective mu-opioid receptor (MOR) phosphorylation. METHODS: In vivo MOR phosphorylation was examined by immunoprecipitation and nanoflow liquid chromatography-tandem mass spectrometry analysis. Using the hot-plate and conditioned place preference test, we investigated opioid-related antinociception and reward effects in mice lacking GRK3 or GRK5. RESULTS: Etonitazene and fentanyl stimulated the in vivo phosphorylation of multiple carboxyl-terminal phosphate acceptor sites, including threonine 370, serine 375, and threonine 379, which was predominantly mediated by GRK3. By contrast, morphine promoted a selective phosphorylation of serine 375 that was predominantly mediated by GRK5. In contrast to GRK3 knockout mice, GRK5 knockout mice exhibited reduced antinociceptive responses after morphine administration and developed morphine tolerance similar to wild-type mice but fewer signs of physical dependence. Also, morphine was ineffective in inducing conditioned place preference in GRK5 knockout mice, whereas cocaine conditioned place preference was retained. However, the reward properties of morphine were evident in knock-in mice expressing a phosphorylation-deficient S375A mutation of the MOR. CONCLUSIONS: These findings show for the first time that MOR phosphorylation is regulated by agonist-selective recruitment of distinct GRK isoforms that influence different opioid-related behaviors. Modulation of GRK5 function could serve as a new approach for preventing addiction to opioids, while maintaining the analgesic properties of opioid drugs at an effective level.


Subject(s)
Analgesics, Opioid/pharmacology , G-Protein-Coupled Receptor Kinase 5/metabolism , Morphine Dependence/enzymology , Morphine/pharmacology , Receptors, Opioid, mu/metabolism , Reward , Animals , Benzimidazoles/pharmacology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Drug Tolerance , Fentanyl/pharmacology , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 3/metabolism , G-Protein-Coupled Receptor Kinase 5/genetics , Gene Knockout Techniques , Mice , Mice, Knockout , Nociception/drug effects , Phosphorylation , Protein Isoforms/drug effects , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics
18.
Bioorg Med Chem Lett ; 22(24): 7471-4, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23131340

ABSTRACT

Based on our earlier reported neuropeptide FF receptors antagonist (RF9), we carried out an extensive structural exploration of the N-terminus part of the amidated dipeptide Arg-Phe-NH(2) in order to establish a structure-activity relationships (SAR) study towards both NPFF receptor subtypes. This SAR led to the discovery of dipeptides (12, 35) with subnanomolar affinities towards NPFF1 receptor subtype, similar to endogenous ligand NPVF. More particularly, compound 12 exhibited a potent in vivo preventive effect on opioid-induced hyperalgesia at low dose. The significant selectivity of 12 toward NPFF1-R indicates that this receptor subtype may play a critical role in the anti-opioid activity of NPFF-like peptides.


Subject(s)
Dipeptides/pharmacology , Receptors, Neuropeptide/antagonists & inhibitors , Dipeptides/chemical synthesis , Dipeptides/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship
19.
PLoS One ; 7(9): e46348, 2012.
Article in English | MEDLINE | ID: mdl-23029489

ABSTRACT

G-protein coupled receptors (GPCRs) play a major role in a number of physiological and pathological processes. Thus, GPCRs have become the most frequent targets for development of new therapeutic drugs. In this context, the availability of highly specific antibodies may be decisive to obtain reliable findings on localization, function and medical relevance of GPCRs. However, the rapid and easy generation of highly selective anti-GPCR antibodies is still a challenge. Herein, we report that highly specific antibodies suitable for detection of GPCRs in native and unfolded forms can be elicited by immunizing animals against purified full length denatured recombinant GPCRs. Contrasting with the currently admitted postulate, our study shows that an active and well-folded GPCR is not required for the production of specific anti-GPCR antibodies. This new immunizing strategy validated with three different human GPCR (µ-opioid, κ-opioid, neuropeptide FF2 receptors) might be generalized to other members of the GPCR family.


Subject(s)
Antibodies/immunology , Immunoglobulin G/biosynthesis , Receptors, Neuropeptide/immunology , Receptors, Opioid, kappa/immunology , Receptors, Opioid, mu/immunology , Amino Acid Sequence , Animals , Antibodies/isolation & purification , Humans , Immunization , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pichia/genetics , Protein Denaturation , Protein Folding , Receptors, Neuropeptide/administration & dosage , Receptors, Neuropeptide/genetics , Receptors, Opioid, kappa/administration & dosage , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/administration & dosage , Receptors, Opioid, mu/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology
20.
Peptides ; 37(1): 157-60, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22813580

ABSTRACT

Neuropeptide FF (NPFF) has been shown to act as an endogenous anti-analgesic peptide. In this paper, several peptide analogs of the selective ligand dNP(NMe)AFLFQPQRF-NH(2) modified in the putative address segment, were designed to be selective NPFF(2) receptor probes, synthesized and assayed. One peptide dA(NMe)AAFLFQPQRF-NH(2) displays a very high affinity for NPFF(2) receptors transfected in CHO cells, and a high selectivity versus NPFF(1) receptors. The exact residues carried in the N-terminal part of the ligands are not decisive to obtain a high affinity only the length of the peptide in itself seems important to create selectivity.


Subject(s)
Peptide Fragments/pharmacology , Receptors, Neuropeptide/agonists , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Humans , Ligands , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Neuropeptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...