Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters










Publication year range
1.
Molecules ; 25(18)2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32957550

ABSTRACT

The opioid receptors are members of the G-protein-coupled receptor (GPCR) family and are known to modulate a variety of biological functions, including pain perception. Despite considerable advances, the mechanisms by which opioid agonists and antagonists interact with their receptors and exert their effect are still not completely understood. In this report, six new hybrids of the Dmt-Tic pharmacophore and cyclic peptides, which were shown before to have a high affinity for the µ-opioid receptor (MOR) were synthesized and characterized pharmacologically in calcium mobilization functional assays. All obtained ligands turned out to be selective antagonists of the δ-opioid receptor (DOR) and did not activate or block the MOR. The three-dimensional structural determinants responsible for the DOR antagonist properties of these analogs were further investigated by docking studies. The results indicate that these compounds attach to the DOR in a slightly different orientation with respect to the Dmt-Tic pharmacophore than Dmt-TicΨ[CH2-NH]Phe-Phe-NH2 (DIPP-NH2[Ψ]), a prototypical DOR antagonist peptide. Key pharmacophoric contacts between the DOR and the ligands were maintained through an analogous spatial arrangement of pharmacophores, which could provide an explanation for the predicted high-affinity binding and the experimentally observed functional properties of the novel synthetic ligands.


Subject(s)
Dipeptides/metabolism , Opioid Peptides/metabolism , Peptides, Cyclic/metabolism , Receptors, Opioid, delta/metabolism , Tetrahydroisoquinolines/metabolism , Analgesics, Opioid/antagonists & inhibitors , Animals , Humans , Ligands , Molecular Docking Simulation , Narcotic Antagonists/chemistry , Narcotic Antagonists/metabolism , Opioid Peptides/chemical synthesis , Peptides, Cyclic/chemical synthesis , Receptors, Opioid/chemistry , Structure-Activity Relationship
2.
Molecules ; 25(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731576

ABSTRACT

In the present contribution, we analyze the influence that C-terminal extension of short opioid peptide sequences by organic fragments has on receptor affinity, in vivo analgesic activity, and antimelanoma properties. The considered fragments were based on either N-acylhydrazone (NAH) or N'-acylhydrazide motifs combined with the 3,5-bis(trifluoromethyl)phenyl moiety. Eleven novel compounds were synthesized and subject to biological evaluation. The analyzed compounds exhibit a diversified range of affinities for the µ opioid receptor (MOR), rather low δ opioid receptor (DOR) affinities, and no appreciable neurokinin-1 receptor binding. In three out of four pairs, N-acylhydrazone-based derivatives bind MOR better than their N'-acylhydrazide counterparts. The best of the novel derivatives have similar low nanomolar MOR binding affinity as the reference opioids, such as morphine and biphalin. The obtained order of MOR affinities was compared to the results of molecular docking. In vivo, four tested compounds turned out to be relatively strong analgesics. Finally, the NAH-based analogues reduce the number of melanoma cells in cell culture, while their N'-acylhydrazide counterparts do not. The antimelanoma properties are roughly correlated to the lipophilicity of the compounds.


Subject(s)
Analgesics , Cytotoxins , Hydrazones/chemistry , Melanoma/drug therapy , Molecular Docking Simulation , Opioid Peptides , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacology , Animals , Cell Line, Tumor , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacology , Humans , Male , Melanoma/metabolism , Melanoma/pathology , Opioid Peptides/chemical synthesis , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Rats , Rats, Wistar , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism
3.
Peptides ; 131: 170369, 2020 09.
Article in English | MEDLINE | ID: mdl-32673700

ABSTRACT

Peptides are an important class of molecules with diverse biological activities. Many endogenous peptides, especially neuropeptides and peptide hormones, play critical roles in development and regulating homeostasis. Furthermore, as drug candidates their high receptor selectivity and potent binding leads to reduced off-target interactions and potential negative side effects. However, the therapeutic potential of peptides is severely hampered by their poor stability in vivo and low permeability across biological membranes. Several strategies have been successfully employed over the decades to address these concerns, and one of the most promising strategies is glycosylation. It has been demonstrated in numerous cases that glycosylation is an effective synthetic approach to improve the pharmacokinetic profiles and membrane permeability of peptides. The effects of glycosylation on peptide stability and peptide-membrane interactions in the context of blood-brain barrier penetration will be explored. Numerous examples of glycosylated analogues of endogenous peptides targeting class A and B G-protein coupled receptors (GPCRs) with an emphasis on O-linked glycopeptides will be reviewed. Notable examples of N-, S-, and C-linked glycopeptides will also be discussed. A small section is devoted to synthetic methods for the preparation of glycopeptides and requisite amino acid glycoside building blocks.


Subject(s)
Biological Products/pharmacology , Blood-Brain Barrier/metabolism , Glycopeptides/pharmacology , Opioid Peptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Amino Acids , Biological Products/isolation & purification , Biological Products/metabolism , Blood-Brain Barrier/drug effects , Central Nervous System/drug effects , Central Nervous System/metabolism , Chemistry Techniques, Synthetic , Glycopeptides/chemical synthesis , Glycopeptides/classification , Glycopeptides/metabolism , Glycosides/chemistry , Glycosides/metabolism , Glycosylation , Humans , Opioid Peptides/chemical synthesis , Opioid Peptides/metabolism , Protein Stability , Proteolysis , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics
4.
Drug Dev Res ; 80(4): 425-437, 2019 06.
Article in English | MEDLINE | ID: mdl-30681179

ABSTRACT

VV-Hemorphin-5 is an endogenous opioid peptide of the Hemorphin family with affinity at opioid receptors. A series of C-amide analogues have been synthesized, based on the structure of VV-Hemorphin-5, modified at position 1 and 7 by the un/natural amino acids (Aa8-Val-Val-Tyr-Pro-Trp-Thr-Gln-NH2 and Val-Val-Tyr-Pro-Trp-Thr-Aa1-NH2 ) using SPPS, Fmoc-chemistry. The peptide derivatives were evaluated for their anticonvulsant activity in three acute seizure tests in male ICR mice, the maximal electroshock (MES), the 6 Hz psychomotor seizure test, and the timed intravenous pentylenetetrazole (ivPTZ) infusion test. Their neurotoxicity was assessed in the rotarod test. Among the tested peptide analogues, V4 showed anticonvulsant activity in the three seizure tests that was comparable to the VV-Hemorphin-5 (V1) used as a positive control. While V5, V6, and V7 peptide derivatives exhibited anticonvulsant activity in the MES and 6 Hz test, they were inactive (V7) or showed pro-convulsant effect (V5 and V6) in the i.v. PTZ test. At a dose of 10 µg/mouse the peptide V2 was effective against clonic seizures induced by PTZ. Motor coordination was not affected by newly developed analogues of VV-Hemorphin-5. Docking study results suggest that kappa opioid receptor binding could be the mechanism of action of peptide derivatives with anticonvulsant activity. The results suggest that incorporation of nonproteinogenic and/or natural amino acids at position 1 and 7 of the VV-Hemorphin-5 scaffold deserve further evaluation in models of epilepsy and derivatization.


Subject(s)
Anticonvulsants/pharmacology , Hemoglobins/pharmacology , Opioid Peptides/pharmacology , Peptide Fragments/pharmacology , Seizures/drug therapy , Amino Acid Sequence , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Electroshock , Hemoglobins/chemical synthesis , Hemoglobins/chemistry , Male , Mice, Inbred ICR , Molecular Docking Simulation , Molecular Structure , Opioid Peptides/chemical synthesis , Opioid Peptides/chemistry , Pentylenetetrazole , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Protein Binding , Psychomotor Performance/drug effects , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Structure-Activity Relationship
5.
Peptides ; 99: 195-204, 2018 01.
Article in English | MEDLINE | ID: mdl-29031796

ABSTRACT

Based on their high selectivity of action and low toxicity, naturally occurring peptides have great potential in terms of drug development. However, the pharmacokinetic properties of peptides, in particular their half life, are poor. Among different strategies developed for reducing susceptibility to peptidases, and thus increasing the duration of action of peptides, the generation of branched peptides has been described. However, the synthesis and purification of branched peptides are extremely complicated thus limiting their druggability. Here we present a novel and facile synthesis of tetrabranched peptides acting as GPCR ligands and their in vitro and vivo pharmacological characterization. Tetrabranched derivatives of nociceptin/orphanin FQ (N/OFQ), N/OFQ related peptides, opioid peptides, tachykinins, and neuropeptide S were generated with the strategy named peptide welding technology (PWT) and characterized by high yield and purity of the desired final product. In general, PWT derivatives displayed a pharmacological profile similar to that of the natural sequence in terms of affinity, pharmacological activity, potency, and selectivity of action in vitro. More importantly, in vivo studies demonstrated that PWT peptides are characterized by increased potency associated with long lasting duration of action. In conclusion, PWT derivatives of biologically active peptides can be viewed as innovative pharmacological tools for investigating those conditions and states in which selective and prolonged receptor stimulation promotes beneficial effects.


Subject(s)
Opioid Peptides , Protein Engineering/methods , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Opioid Peptides/chemical synthesis , Opioid Peptides/chemistry , Opioid Peptides/pharmacology
6.
Biopolymers ; 108(5)2017 Sep.
Article in English | MEDLINE | ID: mdl-28464209

ABSTRACT

To date structure-activity relationship (SAR) studies of the dynorphins (Dyn), endogenous peptides for kappa opioid receptors (KOR), have focused almost exclusively on Dyn A with minimal studies on Dyn B. While both Dyn A and Dyn B have identical N-terminal sequences, their C-terminal sequences differ, which could result in differences in pharmacological activity. We performed an alanine scan of the non-glycine residues up through residue 11 of Dyn B amide to explore the roles of these side chains in the activity of Dyn B. The analogs were synthesized by fluorenylmethyloxycarbonyl (Fmoc)-based solid phase peptide synthesis and evaluated for their opioid receptor affinities and opioid potency and efficacy at KOR. Similar to Dyn A the N-terminal Tyr1 and Phe4 residues of Dyn B amide are critical for opioid receptor affinity and KOR agonist potency. The basic residues Arg6 and Arg7 contribute to the KOR affinity and agonist potency of Dyn B amide, while Lys10 contributes to the opioid receptor affinity, but not KOR agonist potency, of this peptide. Comparison to the Ala analogs of Dyn A (1-13) suggests that the basic residues in the C-terminus of both peptides contribute to KOR binding, but differences in their relative positions may contribute to the different pharmacological profiles of Dyn A and Dyn B. The other unique C-terminal residues in Dyn B amide also appear to influence the relative affinity of this peptide for KOR versus mu and delta opioid receptors. This SAR information may be applied in the design of new Dyn B analogs that could be useful pharmacological tools.


Subject(s)
Alanine/chemistry , Dynorphins/metabolism , Endorphins/metabolism , Opioid Peptides/metabolism , Amides/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Dynorphins/chemical synthesis , Dynorphins/chemistry , Endorphins/chemical synthesis , Endorphins/chemistry , Mutagenesis , Opioid Peptides/chemical synthesis , Opioid Peptides/chemistry , Protein Binding , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Solid-Phase Synthesis Techniques , Structure-Activity Relationship
7.
Sci Rep ; 7: 45817, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28383520

ABSTRACT

The peptide nociceptin/orphanin FQ (N/OFQ) and the N/OFQ receptor (NOP) constitute a neuropeptidergic system that modulates various biological functions and is currently targeted for the generation of innovative drugs. In the present study dimeric NOP receptor ligands with spacers of different lengths were generated using both peptide and non-peptide pharmacophores. The novel compounds (12 peptide and 7 nonpeptide ligands) were pharmacologically investigated in a calcium mobilization assay and in the mouse vas deferens bioassay. Both structure- and conformation-activity studies were performed. Results demonstrated that dimerization did not modify the pharmacological activity of both peptide and non-peptide pharmacophores. Moreover, when dimeric compounds were obtained with low potency peptide pharmacophores, dimerization recovered ligand potency. This effect depends on the doubling of the C-terminal address sequence rather than the presence of an additional N-terminal message sequence or modifications of peptide conformation.


Subject(s)
Dimerization , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Receptors, Opioid/metabolism , Animals , CHO Cells , Calcium/analysis , Cricetulus , Ligands , Opioid Peptides/chemical synthesis , Protein Binding , Protein Multimerization , Nociceptin Receptor , Nociceptin
8.
Mini Rev Med Chem ; 17(6): 486-517, 2017.
Article in English | MEDLINE | ID: mdl-27834131

ABSTRACT

Molecular hybridization is a recent strategy based on the covalent fusion of two or more pharmacophores to create a single molecule with multiple mechanisms of action, which represents an encouraging approach in the development of new drugs with potential therapeutic application in several pathologies. This review provides a comprehensive perspective of the most relevant advances in the development of hybrid molecules acting in the central nervous system. For instance, several opioid hybrids based on endogenous opioid peptides (e.g. enkephalins, deltorphins and endomorphins) have been developed, and γ-aminobutyric acid agonists have also been designed for neuropathic pain control. In addition, a number of hybrid compounds have also been synthesized and evaluated for their anticonvulsant activity and neurotoxicity, which may be further developed as potential antiepileptic drugs. Moreover, several hybrid compounds have also been designed for the treatment of neurodegenerative diseases focusing primarily on Alzheimer's disease by targeting the cholinergic neurotransmission, as acetylcholinesterase inhibitors, and the amyloid ß-protein deposition. There are also studies addressing hybrid compounds including an antioxidant moiety, which can be potentially useful in Alzheimer's and Parkinson's diseases and other neurodegenerative disorders. Additionally, other research works have also shown promising hybrid molecules for depression, autism and cocaine addiction. Thus, the development of molecular hybrid compounds seems to be a promising strategy in the discovery of novel therapeutic drugs.


Subject(s)
Analgesics/chemistry , Central Nervous System Diseases/drug therapy , Opioid Peptides/chemistry , Analgesics/chemical synthesis , Analgesics/therapeutic use , Central Nervous System Diseases/pathology , Choline/chemistry , Donepezil , Epilepsy/drug therapy , Epilepsy/pathology , Humans , Indans/chemistry , Opioid Peptides/chemical synthesis , Opioid Peptides/therapeutic use , Pain/drug therapy , Pain/pathology , Piperidines/chemistry , Tacrine/chemistry , gamma-Aminobutyric Acid/chemistry
9.
J Pept Sci ; 22(8): 545-51, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27443980

ABSTRACT

The synthesis of new dermorphin analogues is described. The (R)-alanine or phenylalanine residues of natural dermorphin were substituted by the corresponding α-methyl-ß-azidoalanine or α-benzyl-ß-azido(1-piperidinyl)alanine residues. The potency and selectivity of the new analogues were evaluated by a competitive receptor binding assay in rat brain using [(3) H]DAMGO (a µ ligand) and [(3) H]DELT (a δ ligand). The most active analogue in this series, Tyr-(R)-Ala-(R)-α-benzyl-ß-azidoAla-Gly-Tyr-Pro-Ser-NH2 and its epimer were analysed by (1) H and (13) C NMR spectroscopy and restrained molecular dynamics simulations. The dominant conformation of the investigated peptides depended on the absolute configuration around C(α) in the α-benzyl-ß-azidoAla residue in position 3. The (R) configuration led to the formation of a type I ß-turn, whilst switching to the (S) configuration gave rise to an inverse ß-turn of type I', followed by the formation of a very short ß-sheet. The selectivity of Tyr-(R)-Ala-(R) and (S)-α-benzyl-ß-azidoAla-Gly-Tyr-Pro-Ser-NH2 was shown to be very similar; nevertheless, the two analogues exhibited different conformational preferences. Copyright © 2016 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Analgesics, Opioid/chemistry , Drug Design , Opioid Peptides/chemistry , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Alanine/chemistry , Amino Acid Sequence , Amino Acid Substitution , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/pharmacology , Animals , Azides/chemistry , Binding Sites , Binding, Competitive , Brain Chemistry , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Ligands , Molecular Dynamics Simulation , Opioid Peptides/chemical synthesis , Opioid Peptides/pharmacology , Phenylalanine/chemistry , Piperidines/chemistry , Protein Binding , Protein Structure, Secondary , Rats , Rats, Wistar , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
10.
J Pept Sci ; 22(4): 222-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27028206

ABSTRACT

In this study, proteinogenic amino acids residues of dimeric dermorphin pentapeptides were replaced by the corresponding ß(3)-homo-amino acids. The potency and selectivity of hybrid α/ß dimeric dermorphin pentapeptides were evaluated by competetive receptor binding assay in the rat brain using [3H]DAMGO (a µ ligand) and [3H]DELT (a δ ligand). Tha analog containing ß(3)-homo-Tyr in place of Tyr (Tyr-D-Ala-Phe-Gly-ß(3)-homo-Tyr-NH-)2 showed good µ receptor affinity and selectivity (IC50 = 0.302, IC50 ratio µ/δ = 68) and enzymatic stability in human plasma.


Subject(s)
Aminobutyrates/chemistry , Opioid Peptides/chemical synthesis , Tyrosine/analogs & derivatives , Tyrosine/chemistry , Animals , Binding, Competitive , Drug Stability , Humans , Inhibitory Concentration 50 , Male , Opioid Peptides/metabolism , Opioid Peptides/pharmacology , Protein Binding , Rats , Rats, Wistar , Receptors, Opioid/metabolism
11.
Bioorg Med Chem ; 22(23): 6545-6551, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25456075

ABSTRACT

Cyclization of linear sequences is a well recognized tool in opioid peptide chemistry for generating analogs with improved bioactivities. Cyclization can be achieved through various bridging bonds between peptide ends or side-chains. In our earlier paper we have reported the synthesis and biological activity of a cyclic peptide, Tyr-c[D-Lys-Phe-Phe-Asp]NH2 (1), which can be viewed as an analog of endomorphin-2 (EM-2, Tyr-Pro-Phe-Phe-NH2). Cyclization was achieved through an amide bond between side-chains of D-Lys and Asp residues. Here, to increase rigidity of the cyclic structure, we replaced d-Lys with cis- or trans-4-aminocyclohexyl-D-alanine (D-ACAla). Two sets of analogs incorporating either Tyr or Dmt (2',6'-dimethyltyrosine) residues in position 1 were synthesized. In the binding studies the analog incorporating Dmt and trans-D-ACAla showed high affinity for both, µ- and δ-opioid receptors (MOR and DOR, respectively) and moderate affinity for the κ-opioid receptor (KOR), while analog with Dmt and cis-D-ACAla was exceptionally MOR-selective. Conformational analyses by NMR and molecular docking studies have been performed to investigate the molecular structural features responsible for the noteworthy MOR selectivity.


Subject(s)
Alanine/analogs & derivatives , Cyclohexanes/chemistry , Opioid Peptides/chemistry , Alanine/chemical synthesis , Alanine/chemistry , Cyclization , Cyclohexanes/chemical synthesis , Humans , Opioid Peptides/chemical synthesis , Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Stereoisomerism
12.
Br J Pharmacol ; 171(17): 4138-53, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24903280

ABSTRACT

BACKGROUND AND PURPOSE: An innovative chemical approach, named peptide welding technology (PWT), allows the synthesis of multibranched peptides with extraordinary high yield, purity and reproducibility. With this approach, three different tetrabranched derivatives of nociceptin/orphanin FQ (N/OFQ) have been synthesized and named PWT1-N/OFQ, PWT2-N/OFQ and PWT3-N/OFQ. In the present study we investigated the in vitro and in vivo pharmacological profile of PWT N/OFQ derivatives and compared their actions with those of the naturally occurring peptide. EXPERIMENTAL APPROACH: The following in vitro assays were used: receptor and [(35)S]-GTPγS binding, calcium mobilization in cells expressing the human N/OFQ peptide (NOP) receptor, or classical opioid receptors and chimeric G proteins, electrically stimulated mouse vas deferens bioassay. In vivo experiments were performed; locomotor activity was measured in normal mice and in animals with the NOP receptor gene knocked out [NOP(-/-)]. KEY RESULTS: In vitro PWT derivatives of N/OFQ behaved as high affinity potent and rather selective full agonists at human recombinant and animal native NOP receptors. In vivo PWT derivatives mimicked the inhibitory effects exerted by the natural peptide on locomotor activity showing 40-fold higher potency and extremely longer lasting action. The effects of PWT2-N/OFQ were no longer evident in NOP(-/-) mice. CONCLUSIONS AND IMPLICATIONS: The results showed that the PWT can be successfully applied to the peptide sequence of N/OFQ to generate tetrabranched derivatives characterized by a pharmacological profile similar to the native peptide and associated with a higher potency and marked prolongation of action in vivo.


Subject(s)
Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Receptors, Opioid/agonists , Animals , CHO Cells , Cells, Cultured , Cricetulus , Dose-Response Relationship, Drug , Humans , Locomotion/drug effects , Male , Mice , Mice, Knockout , Molecular Conformation , Opioid Peptides/chemical synthesis , Receptors, Opioid/deficiency , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 23(15): 4347-50, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23796454

ABSTRACT

Standard radioiodination methods lack site-selectivity and either mask charges (Bolton-Hunter) or involve oxidative reaction conditions (chloramine-T). Opioid peptides are very sensitive to certain structural modifications, making these labeling methods untenable. In our model opioid peptide, α-neoendorphin, we replaced a tyrosyl hydroxyl with an iodine, and in cell lines stably expressing mu, delta, or kappa opioid receptors, we saw no negative effects on binding. We then optimized a repurposed Sandmeyer reaction using copper(I) catalysts with non-redoxing/non-nucleophilic ligands, bringing the radiochemical yield up to around 30%, and site-selectively incorporated radioactive iodine into this position under non-oxidizing reaction conditions, which should be broadly compatible with most peptides. The (125)I- and (131)I-labeled versions of the compound bound with high affinity to opioid receptors in mouse brain homogenates, thus demonstrating the general utility of the labeling strategy and of the peptide for exploring opioid binding sites.


Subject(s)
Endorphins/metabolism , Opioid Peptides/chemistry , Protein Precursors/metabolism , Amino Acid Sequence , Animals , Binding Sites , Brain/metabolism , CHO Cells , Catalysis , Copper/chemistry , Cricetinae , Cricetulus , Endorphins/chemistry , Halogenation , Iodine Radioisotopes/chemistry , Mice , Opioid Peptides/chemical synthesis , Opioid Peptides/metabolism , Protein Binding , Protein Precursors/chemistry , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism
14.
Bioorg Med Chem Lett ; 23(11): 3434-7, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23623418

ABSTRACT

We report here the design, synthesis, and in vitro characterization of new opioid peptides featuring a 4-anilidopiperidine moiety. Despite the fact that the chemical structures of fentanyl surrogates have been found suboptimal per se for the opioid activity, the corresponding conjugates with opioid peptides displayed potent opioid activity. These studies shed an instructive light on the strategies and potential therapeutic values of anchoring the 4-anilidopiperidine scaffold to different classes of opioid peptides.


Subject(s)
Opioid Peptides/chemistry , Piperidines/chemistry , Animals , Cell Line , Fentanyl/chemistry , Humans , Kinetics , Mice , Opioid Peptides/chemical synthesis , Opioid Peptides/metabolism , Piperidines/chemical synthesis , Piperidines/metabolism , Protein Binding , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
15.
Curr Drug Targets ; 14(7): 798-816, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23621510

ABSTRACT

Endogenous opioid peptides have been studied extensively as potential therapeutics for the treatment of pain. The major problems of using natural opioid peptides as drug candidates are their poor receptor specificity, metabolic instability and inability to reach the brain after systemic administration. A lot of synthetic efforts have been made to opioid analogs with improved pharmacological properties. One important structural modification leading to such analogs is cyclization of linear sequences. Intramolecular cyclization has been shown to improve biological properties of various bioactive peptides. Cyclization reduces conformational freedom responsible for the simultaneous activation of two or more receptors, increases metabolic stability and lipophilicity which may result in a longer half-life and easier penetration across biological membranes. This review deals with various strategies that have been employed to synthesize cyclic analogs of opioid peptides. Discussed are such bridging bonds as amide and amine linkages, sulfur-containing bonds, including monosulfide, disulfide and dithioether bridges, bismethylene bonds, monosulfide bridges of lanthionine and, finally, carbonyl and guanidine linkages. Opioid affinities and activities of cyclic analogs are given and compared with linear opioid peptides. Analgesic activities of analogs evaluated in the in vivo pain tests are also discussed.


Subject(s)
Analgesics, Opioid/chemical synthesis , Opioid Peptides/chemical synthesis , Pain/drug therapy , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Brain/metabolism , Cyclization , Half-Life , Humans , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Tissue Distribution
16.
Eur J Med Chem ; 63: 457-67, 2013 May.
Article in English | MEDLINE | ID: mdl-23523659

ABSTRACT

A series of novel cyclic ureidopeptides, analogues of dermorphine/deltorphine tetrapeptide, were synthesized by solid phase peptide synthesis and/or in solution. The antinociceptive activity of N-substituted amides 1-10 was evaluated using hot-plate and tail-flick tests. Analogue 1 showed significant, stronger than morphine, antinociceptive effect after systemic applications. All analogues were also tested for their in vitro resistance to proteolysis by means of mass spectroscopy and it was found that all substituted amides 1-10 showed full stability during incubation with large excess of chymotrypsin and pepsin. Compound 1 is a lead molecule for further evaluation.


Subject(s)
Analgesics, Opioid/chemical synthesis , Hyperalgesia/physiopathology , Oligopeptides/chemical synthesis , Opioid Peptides/chemical synthesis , Analgesics, Opioid/chemistry , Analgesics, Opioid/pharmacology , Animals , Chymotrypsin/metabolism , Hot Temperature/adverse effects , Hydrolysis , Hyperalgesia/etiology , Hyperalgesia/prevention & control , Indoles , Male , Mice , Mice, Inbred BALB C , Models, Chemical , Molecular Structure , Oligopeptides/chemistry , Oligopeptides/pharmacology , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Pepsin A/metabolism , Proteolysis , Spectrometry, Mass, Electrospray Ionization , Styrenes
17.
J Pept Sci ; 19(4): 233-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23136069

ABSTRACT

AM94 is a fluorinated analog of biphalin with non-hydrazine linker that has an in vitro affinity for µ-opioid and δ-opioid receptors tenfold higher than biphalin. Furthermore, in vivo evaluation in rats showed that AM94 has in hot plate test - after both intracerebroventricular and intravenous administrations - a greater and more durable efficacy than biphalin. Here, the antinociceptive profile of AM94 is further evaluated by following two different administration routes, intrathecal and subcutaneous, and two different animal species, rats and mice. The analgesic potency of AM94 is compared with that of both the parent peptide biphalin and morphine. Results show that in rats (tail flick test) and in mice (formalin test), AM94 has a higher and more durable analgesic effect than biphalin after intrathecal and subcutaneous administrations. Conformational properties of biphalin and AM94 were also investigated by variable-temperature (1)H NMR and energy minimization.


Subject(s)
Analgesics , Opioid Peptides , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacology , Animals , Enkephalins/chemistry , Enkephalins/pharmacology , Male , Mice , Morphine/chemistry , Morphine/pharmacology , Opioid Peptides/chemical synthesis , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Protein Structure, Secondary , Rats , Rats, Wistar , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism
18.
Regul Pept ; 179(1-3): 23-8, 2012 Nov 10.
Article in English | MEDLINE | ID: mdl-22960407

ABSTRACT

A chimeric opioid peptide (MCRT, YPFPFRTic-NH(2)) was here designed and synthesized. This peptide was based on morphiceptin (YPFP-NH(2)) and a neuropeptide FF (NPFF) derivative (PFRTic-NH(2)) sharing one proline. This peptide is intended to produce potent analgesia. MCRT was found to induce analgesic activity in a dose- and time-dependent manner, as indicated by a tail flick latency test in mice to which it had been intracerebroventricularly administered (5-60 min, 0.025-2.5 nmol/kg (0.5-50 pmol per mouse), ED(50)=1.49 nmol/kg). At 2.5nmol/kg, MCRT showed significantly higher levels of analgesic activity than morphiceptin or PFR(Tic)amide at 2500 nmol/kg. Naltrindole and cyprodime were found to partially but significantly inhibit this analgesic activity, but naloxone blocked it completely. The kappa opioid receptor antagonist nor-BNI was found to slightly inhibit MCRT and morphiceptin. Pre-injection of BIBP3226 and co-administration of NPFF and MCRT showed that NPFF receptors were involved in the analgesia of MCRT. BIBP3226 was found to weaken the analgesic effects of MCRT, but BIBP3226 could not block the analgesic effects of PFR(Tic)amide. Overall, MCRT was found to have stronger analgesic activity than morphiceptin or PFR(Tic)amide when interacting with mixed µ/δ opioid receptor interactions. MCRT also showed partial interaction with NPFF receptors.


Subject(s)
Analgesics, Opioid/pharmacology , Endorphins/pharmacology , Neuropeptides/pharmacology , Opioid Peptides/pharmacology , Receptors, Neuropeptide/metabolism , Tetrahydroisoquinolines/pharmacology , Analgesia/methods , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/chemical synthesis , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Dose-Response Relationship, Drug , Endorphins/administration & dosage , Endorphins/antagonists & inhibitors , Guinea Pigs , Male , Mice , Morphinans/pharmacology , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Neuropeptides/administration & dosage , Neuropeptides/metabolism , Opioid Peptides/administration & dosage , Opioid Peptides/chemical synthesis , Proline/metabolism , Receptors, Neuropeptide/antagonists & inhibitors , Tetrahydroisoquinolines/administration & dosage , Tetrahydroisoquinolines/metabolism , Time Factors
19.
J Pept Sci ; 18(9): 556-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22786725

ABSTRACT

ß-Amino acids containing α,ß-hybrid peptides show great potential as peptidomimetics. In this paper, we describe the synthesis and affinity to µ-opioid and δ-opioid receptors of α,ß-hybrids, analogs of the tetrapeptide Tyr- d-Ala-Phe-Phe-NH(2) (TAPP). Each amino acid was replaced with an l- or d-ß(3) -h-amino acid. All α,ß-hybrids of TAPP analogs were synthesized in solution and tested for affinity to µ-opioid and δ-opioid receptors. The analog Tyr-ß(3) h- d-Ala-Phe-PheNH(2) was found to be as active as the native tetrapeptide.


Subject(s)
Amino Acids/chemistry , Oligopeptides/chemistry , Opioid Peptides/chemical synthesis , Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Opioid Peptides/chemistry , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...