Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
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
2.
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
3.
J Biol Chem ; 287(16): 12736-49, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22375000

ABSTRACT

Neuropeptide FF (NPFF) interacts with specific receptors to modulate opioid functions in the central nervous system. On dissociated neurons and neuroblastoma cells (SH-SY5Y) transfected with NPFF receptors, NPFF acts as a functional antagonist of µ-opioid (MOP) receptors by attenuating the opioid-induced inhibition of calcium conductance. In the SH-SY5Y model, MOP and NPFF(2) receptors have been shown to heteromerize. To understand the molecular mechanism involved in the anti-opioid activity of NPFF, we have investigated the phosphorylation status of the MOP receptor using phospho-specific antibody and mass spectrometry. Similarly to direct opioid receptor stimulation, activation of the NPFF(2) receptor by [D-Tyr-1-(NMe)Phe-3]NPFF (1DMe), an analog of NPFF, induced the phosphorylation of Ser-377 of the human MOP receptor. This heterologous phosphorylation was unaffected by inhibition of second messenger-dependent kinases and, contrarily to homologous phosphorylation, was prevented by inactivation of G(i/o) proteins by pertussis toxin. Using siRNA knockdown we could demonstrate that 1DMe-induced Ser-377 cross-phosphorylation and MOP receptor loss of function were mediated by the G protein receptor kinase GRK2. In addition, mass spectrometric analysis revealed that the phosphorylation pattern of MOP receptors was qualitatively similar after treatment with the MOP agonist Tyr-D-Ala-Gly (NMe)-Phe-Gly-ol (DAMGO) or after treatment with the NPFF agonist 1DMe, but the level of multiple phosphorylation was more intense after DAMGO. Finally, NPFF(2) receptor activation was sufficient to recruit ß-arrestin2 to the MOP receptor but not to induce its internalization. These data show that NPFF-induced heterologous desensitization of MOP receptor signaling is mediated by GRK2 and could involve transphosphorylation within the heteromeric receptor complex.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Opioid, mu/metabolism , Amino Acid Sequence , Analgesics, Opioid/pharmacology , Arrestins/metabolism , Cell Line, Tumor , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , G-Protein-Coupled Receptor Kinase 2/genetics , Gene Knockdown Techniques , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Neuroblastoma , Phosphorylation/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, mu/agonists , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Serine/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , beta-Arrestins
4.
Anal Biochem ; 420(1): 99-100, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21951781

ABSTRACT

Functional and structural studies on G-protein-coupled receptors (GPCRs) at molecular levels require producing and purifying high levels of receptors, and recombinant mammalian cell expression systems constitute the best systems to obtain receptors resembling those expressed in natural environments. In the course of increasing GPCR expression in Chinese hamster ovary (CHO) cells, we have expressed mu (µ)- and kappa (κ)-opioid receptors and neuropeptide FF(1) and FF(2) receptors (NPFF(1) and NPFF(2), respectively) in dimethyl sulfoxide. This treatment did not modify the affinity (K(d)) for any receptor, but a significant increase in functional expression levels was observed for all receptors with the noticeable exception of NPFF(1).


Subject(s)
Dimethyl Sulfoxide/pharmacology , Receptors, Neuropeptide/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Recombinant Proteins/metabolism , Animals , CHO Cells/drug effects , Cricetinae , Cricetulus , Female , Protein Engineering/methods , Receptors, Neuropeptide/genetics , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/genetics , Recombinant Proteins/genetics
5.
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
6.
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
7.
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
8.
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
9.
Anal Biochem ; 398(2): 225-9, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20018158

ABSTRACT

Neuropeptide FF (NPFF, FLFQPQRFamide) receptors modulate endogenous opioid functions. Here, we report the solubilization of the human NPFF(2) receptor expressed in Chinese hamster ovary (CHO) cells by the zwitterionic detergent Chaps. Chaps solubilization resulted in the abolishment of specific agonist binding activity, which was restored by a polyethylene glycol (PEG) precipitation method. Reincorporation after the precipitation step into liposomes made of endogenous lipids issued from CHO membranes or exogenous lipids significantly enhanced the specific agonist binding activity and G-protein coupling. This method of solubilization and lipid reconstitution could be useful for studies of NPFF receptors.


Subject(s)
Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Animals , CHO Cells , Cholic Acids/pharmacology , Cricetinae , Cricetulus , Detergents/pharmacology , Gene Expression/drug effects , Humans , Receptors, Neuropeptide/genetics , Solubility
10.
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
11.
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
12.
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
13.
Biochem Biophys Res Commun ; 373(1): 80-4, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18544342

ABSTRACT

Mu opioid (MOP) receptor activation can be functionally modulated by stimulation of Neuropeptide FF 2 (NPFF(2)) G protein-coupled receptors. Fluorescence recovery after photobleaching experiments have shown that activation of the NPFF(2) receptor dramatically reduces the fraction of MOP receptors confined in microdomains of the plasma membrane of SH-SY5Y neuroblastoma cells. The aim of the present work was to assess if the direct observation of receptor compartmentation by fluorescence techniques in living cells could be related to indirect estimation of receptor partitioning in lipid rafts after biochemical fractionation of the cell. Our results show that MOP receptor distribution in lipid rafts is highly dependent upon the method of purification, questioning the interpretation of previous data regarding MOP receptor compartmentation. Moreover, the NPFF analogue 1DMe does not modify the distribution profile of MOP receptors, clearly demonstrating that membrane fractionation data do not correlate with direct measurement of receptor compartmentation in living cells.


Subject(s)
Membrane Microdomains/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Opioid, mu/metabolism , Cell Fractionation , Cell Line , Detergents/chemistry , Fluorescence Recovery After Photobleaching , Humans , Oligopeptides/pharmacology , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics
14.
Peptides ; 28(5): 1125-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17418451

ABSTRACT

Activation of the NPFF(2) receptor reduces the inhibitory effect of opioids on the N-type Ca(2+) channel. Although this anti-opioid effect is specific for opioid receptors in neurons and tissues, it also affects NPY Y2 and alpha(2)-adrenoreceptors in undifferentiated SH-SY5Y cells stably expressing the NPFF(2) receptor. To test whether this difference could be due to the immaturity of these cells, they were differentiated to a noradrenergic neuronal phenotype with staurosporine. The differentiated cells ceased to divide and grew long, thin neurites. The inhibition of the depolarization-triggered Ca(2+) transient by activation of G(i)-coupled receptors was either unaffected (micro-opioid), increased (NPY), reduced (NPFF(2)) or lost (alpha(2)-adrenoreceptors). Following a 20 min incubation with 1DMe, the effect of DAMGO was reduced, as in undifferentiated cells, but the effect of NPY was no longer affected. Staurosporine differentiation did not modify the coupling of the micro-opioid and NPFF(2) receptors to the G(i/o) proteins. We suggest that the specificity of the effect of NPFF may not reside in the molecular mechanism of its anti-opioid activity itself but in the organization of receptors within the membrane.


Subject(s)
Cell Differentiation/drug effects , Oligopeptides/metabolism , Receptors, Neuropeptide/physiology , Receptors, Opioid/metabolism , Staurosporine/pharmacology , Calcium/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Clonidine/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Humans , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuropeptide Y/pharmacology , Oligopeptides/pharmacology , Receptors, Neuropeptide/genetics
15.
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
16.
Peptides ; 27(5): 980-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16488058

ABSTRACT

In order to elucidate the mechanisms of the neuronal anti-opioid activity of Neuropeptide FF, we have transfected the SH-SY5Y neuroblastoma cell line, which expresses mu- and delta-opioid receptors, with the human NPFF1 receptor. The SH1-C7 clone expresses high affinity NPFF1 receptors in the same range order of density as opioid receptors. Similarly to the opioids, acute stimulation with the NPFF1 agonist NPVF inhibits adenylyl cyclase activity and voltage-gated (N-type) Ca2+ currents and enhances the intracellular Ca2+ release triggered by muscarinic receptors activation. In contrast, preincubation of cells with NPVF decreases the response to opioids on both calcium signaling, thus reproducing the cellular anti-opioid activity described in neurons. SH1-C7 cells are therefore a suitable model to investigate the interactions between NPFF and opioid receptors.


Subject(s)
Narcotic Antagonists , Receptors, Neuropeptide/physiology , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/physiology , Carbachol/pharmacology , Cell Line, Tumor , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Humans , Neuroblastoma , Neuropeptides/pharmacology , Oligopeptides/pharmacology , Pertussis Toxin/pharmacology , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/physiology , Receptors, Opioid, mu/agonists , Signal Transduction/drug effects , Transfection
17.
Peptides ; 27(5): 990-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16490282

ABSTRACT

Neuropeptide FF, a member of the RFamide family of peptides, has demonstrated an interesting array of pharmacological effects. To date however, little information has been obtained as to the exact pharmacological roles of the individual NPFF1 and NPFF2 receptors. Through peptide analogs of NPFF and related peptides, the essential pharmacophore has emerged somewhat. Yet, the field is lacking small molecule ligands selective for each receptor. This review of the structure-activity relationships of the reported NPFF peptide analogs and some non-selective small molecule ligands highlights the current understanding of the pharmacophoric elements required for affinity and activity at the NPFF receptors. The lack of mutagenesis data on the receptor as well as a crystal structure has also hindered the understanding of ligand recognition at the receptor level. If the targets can be further investigated as to their requirements for ligand recognition, the successful development of highly selective ligands should follow.


Subject(s)
Oligopeptides/physiology , Amino Acid Sequence , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , CHO Cells , Cricetinae , Cricetulus , Humans , Peptide Fragments , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/physiology , Structure-Activity Relationship
18.
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
19.
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
20.
Curr Top Med Chem ; 5(3): 341-55, 2005.
Article in English | MEDLINE | ID: mdl-15857316

ABSTRACT

Opioids are involved in the physiological control of numerous functions of the central nervous system, particularly nociception. It appears that some endogenous neuropeptides, called anti-opioids, participate in an homeostatic system tending to reduce the effects of opioids. Neuropeptide FF (NPFF) and cholecystokinin (CCK) possess these properties and, paradoxically, the opioid peptides nociceptin and dynorphin display some anti-opioid activity. All these peptides exhibit complex properties as they are able to both counteract and potentiate opioid activity, acting rather as modulators of opioid functions. The purpose of this review is to highlight that two different mechanisms are clearly involved in the control of opioid functions by opioid-modulating peptides: a circuitry-induced mechanism for nociceptin and dynorphin, and a cellular anti-opioid mechanism for NPFF and CCK. The knowledge of these mechanisms has potential therapeutic interest in the control of opioid functions, notably for alleviating pain and/or for the treatment of opioid abuse.


Subject(s)
Analgesics, Opioid/pharmacology , Homeostasis/drug effects , Neurons/drug effects , Opioid Peptides/pharmacology , Receptors, Opioid/metabolism , Amino Acid Sequence , Cholecystokinin/metabolism , Dynorphins/metabolism , Homeostasis/physiology , Molecular Sequence Data , Morphine/pharmacology , Neurons/metabolism , Oligopeptides/metabolism , Opioid Peptides/metabolism , Nociceptin
SELECTION OF CITATIONS
SEARCH DETAIL