Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Molecules ; 27(24)2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36557917

ABSTRACT

There is still an unmet clinical need to develop new pharmaceuticals for effective and safe pain management. Current pharmacotherapy offers unsatisfactory solutions due to serious side effects related to the chronic use of opioid drugs. Prescription opioids produce analgesia through activation of the mu-opioid receptor (MOR) and are major contributors to the current opioid crisis. Multifunctional ligands possessing activity at more than one receptor represent a prominent therapeutic approach for the treatment of pain with fewer adverse effects. We recently reported on the design of a bifunctional MOR agonist/neuropeptide FF receptor (NPFFR) antagonist peptididomimetic, KGFF09 (H-Dmt-DArg-Aba-ßAla-Bpa-Phe-NH2), and its antinociceptive effects after subcutaneous (s.c.) administration in acute and persistent pain in mice with reduced propensity for unwanted side effects. In this study, we further investigated the antinociceptive properties of KGFF09 in a mouse model of visceral pain after s.c. administration and the potential for opioid-related liabilities of rewarding and sedation/locomotor dysfunction following chronic treatment. KGFF09 produced a significant dose-dependent inhibition of the writhing behavior in the acetic acid-induced writhing assay with increased potency when compared to morphine. We also demonstrated the absence of harmful effects caused by typical MOR agonists, i.e., rewarding effects (conditioned-place preference test) and sedation/locomotor impairment (open-field test), at a dose shown to be highly effective in inhibiting pain behavior. Consequently, KGFF09 displayed a favorable benefit/side effect ratio regarding these opioid-related side effects compared to conventional opioid analgesics, such as morphine, underlining the development of dual MOR agonists/NPFFR antagonists as improved treatments for various pain conditions.


Subject(s)
Peptidomimetics , Visceral Pain , Mice , Animals , Analgesics, Opioid , Peptidomimetics/pharmacology , Visceral Pain/drug therapy , Visceral Pain/chemically induced , Morphine/pharmacology , Receptors, Opioid, mu/metabolism , GTP-Binding Proteins
2.
Molecules ; 26(11)2021 05 28.
Article in English | MEDLINE | ID: mdl-34071603

ABSTRACT

Opioids are the most effective analgesics, with most clinically available opioids being agonists to the µ-opioid receptor (MOR). The MOR is also responsible for their unwanted effects, including reward and opioid misuse leading to the current public health crisis. The imperative need for safer, non-addictive pain therapies drives the search for novel leads and new treatment strategies. In this study, the recently discovered MOR/nociceptin (NOP) receptor peptide hybrid KGNOP1 (H-Dmt-D-Arg-Aba-ß-Ala-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2) was evaluated following subcutaneous administration in mouse models of acute (formalin test) and chronic inflammatory pain (Complete Freund's adjuvant-induced paw hyperalgesia), liabilities of spontaneous locomotion, conditioned place preference, and the withdrawal syndrome. KGNOP1 demonstrated dose-dependent antinociceptive effects in the formalin test, and efficacy in attenuating thermal hyperalgesia with prolonged duration of action. Antinociceptive effects of KGNOP1 were reversed by naltrexone and SB-612111, indicating the involvement of both MOR and NOP receptor agonism. In comparison with morphine, KGNOP1 was more potent and effective in mouse models of inflammatory pain. Unlike morphine, KGNOP1 displayed reduced detrimental liabilities, as no locomotor impairment nor rewarding and withdrawal effects were observed. Docking of KGNOP1 to the MOR and NOP receptors and subsequent 3D interaction pattern analyses provided valuable insights into its binding mode. The mixed MOR/NOP receptor peptide KGNOP1 holds promise in the effort to develop new analgesics for the treatment of various pain states with fewer MOR-mediated side effects, particularly abuse and dependence liabilities.


Subject(s)
Oligopeptides/genetics , Opioid Peptides/chemistry , Receptors, Opioid, mu/metabolism , Acute Pain/drug therapy , Analgesics , Animals , Behavior, Animal , CHO Cells , Cricetinae , Cricetulus , Cycloheptanes/pharmacology , Humans , Hyperalgesia/drug therapy , In Vitro Techniques , Inflammation/drug therapy , Male , Mice , Models, Molecular , Molecular Docking Simulation , Morphine/chemistry , Morphine/pharmacology , Movement/drug effects , Naloxone/pharmacology , Naltrexone/pharmacology , Pain Management , Piperidines/pharmacology , Nociceptin
3.
J Med Chem ; 63(21): 12929-12941, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32902268

ABSTRACT

Fusion of nonopioid pharmacophores, such as neurotensin, with opioid ligands represents an attractive approach for pain treatment. Herein, the µ-/δ-opioid agonist tetrapeptide H-Dmt-d-Arg-Aba-ß-Ala-NH2 (KGOP01) was fused to NT(8-13) analogues. Since the NTS1 receptor has been linked to adverse effects, selective MOR-NTS2 ligands are preferred. Modifications were introduced within the native NT sequence, particularly a ß3-homo amino acid in position 8 and Tyr11 substitutions. Combination of ß3hArg and Dmt led to peptide 7, a MOR agonist, showing the highest NTS2 affinity described to date (Ki = 3 pM) and good NTS1 affinity (Ki = 4 nM), providing a >1300-fold NTS2 selectivity. The (6-OH)Tic-containing analogue 9 also exhibited high NTS2 affinity (Ki = 1.7 nM), with low NTS1 affinity (Ki = 4.7 µM), resulting in an excellent NTS2 selectivity (>2700). In mice, hybrid 7 produced significant and prolonged antinociception (up to 8 h), as compared to the KGOP01 opioid parent compound.


Subject(s)
Drug Design , Peptides/chemistry , Receptors, Neurotensin/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Amino Acid Sequence , Animals , Disease Models, Animal , Humans , Male , Mice , Oligopeptides/chemistry , Oligopeptides/metabolism , Oligopeptides/therapeutic use , Pain/drug therapy , Pain/pathology , Peptides/metabolism , Peptides/therapeutic use , Protein Binding , Receptors, Neurotensin/chemistry , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Structure-Activity Relationship
4.
Molecules ; 25(9)2020 Apr 29.
Article in English | MEDLINE | ID: mdl-32365707

ABSTRACT

The mu opioid receptor (MOR) is the primary target for analgesia of endogenous opioid peptides, alkaloids, synthetic small molecules with diverse scaffolds, and peptidomimetics. Peptide-based opioids are viewed as potential analgesics with reduced side effects and have received constant scientific interest over the years. This study focuses on three potent peptide and peptidomimetic MOR agonists, DALDA, [Dmt1]DALDA, and KGOP01, and the prototypical peptide MOR agonist DAMGO. We present the first molecular modeling study and structure-activity relationships aided by in vitro assays and molecular docking of the opioid peptide analogues, in order to gain insight into their mode of binding to the MOR. In vitro binding and functional assays revealed the same rank order with KGOP01 > [Dmt1]DALDA > DAMGO > DALDA for both binding and MOR activation. Using molecular docking at the MOR and three-dimensional interaction pattern analysis, we have rationalized the experimental outcomes and highlighted key amino acid residues responsible for agonist binding to the MOR. The Dmt (2',6'-dimethyl-L-Tyr) moiety of [Dmt1]DALDA and KGOP01 was found to represent the driving force for their high potency and agonist activity at the MOR. These findings contribute to a deeper understanding of MOR function and flexible peptide ligand-MOR interactions, that are of significant relevance for the future design of opioid peptide-based analgesics.


Subject(s)
Oligopeptides/chemistry , Oligopeptides/metabolism , Peptides/chemistry , Peptides/metabolism , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Animals , Binding Sites , CHO Cells , Cricetulus , Humans , Kinetics , Mice , Models, Molecular , Protein Binding , Protein Conformation , Structure-Activity Relationship
5.
Sci Rep ; 10(1): 5653, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32221355

ABSTRACT

Morphine and structurally-derived compounds are µ opioid receptor (µOR) agonists, and the most effective analgesic drugs. However, their usefulness is limited by serious side effects, including dependence and abuse potential. The N-substituent in morphinans plays an important role in opioid activities in vitro and in vivo. This study presents the synthesis and pharmacological evaluation of new N-phenethyl substituted 14-O-methylmorphinan-6-ones. Whereas substitution of the N-methyl substituent in morphine (1) and oxymorphone (2) by an N-phenethyl group enhances binding affinity, selectivity and agonist potency at the µOR of 1a and 2a, the N-phenethyl substitution in 14-methoxy-N-methylmorphinan-6-ones (3 and 4) converts selective µOR ligands into dual µ/δOR agonists (3a and 4a). Contrary to N-methylmorphinans 1-4, the N-phenethyl substituted morphinans 1a-4a produce effective and potent antinociception without motor impairment in mice. Using docking and molecular dynamics simulations with the µOR, we establish that N-methylmorphinans 1-4 and their N-phenethyl counterparts 1a-4a share several essential receptor-ligand interactions, but also interaction pattern differences related to specific structural features, thus providing a structural basis for their pharmacological profiles. The emerged structure-activity relationships in this class of morphinans provide important information for tuning in vitro and in vivo opioid activities towards discovery of effective and safer analgesics.


Subject(s)
Analgesics, Opioid/pharmacology , Morphinans/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Humans , Ligands , Male , Mice , Morphine/pharmacology , Structure-Activity Relationship
6.
Pain ; 160(12): 2724-2742, 2019 12.
Article in English | MEDLINE | ID: mdl-31365471

ABSTRACT

About half of patients with spinal cord injury (SCI) develop debilitating central neuropathic pain (CNP), with no effective treatments. Thus, effective, safe, and novel therapies are needed urgently. Previously, docosahexaenoic acid (DHA) was reported to confer neuroprotection in preclinical SCI models. However, its therapeutic potential on SCI-CNP remains to be elucidated. Here, we demonstrated for the first time that intravenous DHA administrations with 3-day intervals (250 nmol/kg; starting 30 minutes after injury and maintained for 6 weeks) effectively prevented SCI-CNP development in a clinically relevant rat contusion model. SCI-CNP was assessed by a novel sensory profiling approach combining evoked pain measures and pain-related ethologically relevant rodent behaviours (burrowing, thigmotaxis, and place/escape avoidance) to mimic those for measuring human (sensory, affective, cognitive, and spontaneous) pain. Strikingly, already established SCI-CNP could be abolished partially by similar DHA administrations, starting from the beginning of week 4 after injury and maintained for 4 weeks. At spinal (epicenter and L5 dorsal horns) and supraspinal (anterior cingulate cortex) levels, both treatment regimens potently suppressed microglial and astrocyte activation, which underpins SCI-CNP pathogenesis. Spinal microgliosis, a known hallmark associated with neuropathic pain behaviours, was reduced by DHA treatments. Finally, we revealed novel potential roles of peroxisome proliferator-activated and retinoid X receptors and docosahexaenoyl ethanolamide (DHA's metabolite) in mediating DHA's effects on microglial activation. Our findings, coupled with the excellent long-term clinical safety of DHA even in surgical and critically ill patients, suggest that systemic DHA treatment is a translatable, effective, safe, and novel approach for preventing and managing SCI-CNP.


Subject(s)
Docosahexaenoic Acids/therapeutic use , Hyperalgesia/prevention & control , Microglia/drug effects , Motor Activity/drug effects , Neuralgia/prevention & control , Spinal Cord Injuries/complications , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Hyperalgesia/etiology , Hyperalgesia/metabolism , Male , Microglia/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Pain Measurement , Rats , Rats, Wistar , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Injuries/metabolism
7.
J Med Chem ; 61(21): 9784-9789, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30351003

ABSTRACT

Chronic pain is currently treated with opioids that offer unsatisfactory long-term analgesia and produce serious side effects. There is a clear need for alternative therapies. Herein, peptide-based hydrogels are used as extended-release drug delivery carriers. Two different formulations were developed: the drug is coformulated within the hydrogel; the drug is an integral part of the hydrogelator. Both strategies afford a prolonged and significant antinociception up to 72 h after subcutaneous administration in mice.


Subject(s)
Drug Carriers/chemistry , Hydrogels/chemistry , Hydrophobic and Hydrophilic Interactions , Oligopeptides/chemistry , Opioid Peptides/chemistry , Opioid Peptides/pharmacology , Amino Acid Sequence , Animals , Drug Carriers/metabolism , Drug Stability , Humans , Mice , Oligopeptides/metabolism
8.
Pain ; 159(9): 1705-1718, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29708942

ABSTRACT

Opioid analgesics, such as morphine, oxycodone, and fentanyl, are the cornerstones for treating moderate to severe pain. However, on chronic administration, their efficiency is limited by prominent side effects such as analgesic tolerance and dependence liability. Neuropeptide FF (NPFF) and its receptors (NPFF1R and NPFF2R) are recognized as an important pronociceptive system involved in opioid-induced hyperalgesia and analgesic tolerance. In this article, we report the design of multitarget peptidomimetic compounds that show high-affinity binding to the mu-opioid receptor (MOPr) and NPFFRs. In vitro characterization of these compounds led to identification of KGFF03 and KGFF09 as G-protein-biased MOPr agonists with full agonist or antagonist activity at NPFFRs, respectively. In agreement with their biased MOPr agonism, KGFF03/09 showed reduced respiratory depression in mice, as compared to the unbiased parent opioid agonist KGOP01. Chronic subcutaneous administration of KGOP01 and KGFF03 in mice rapidly induced hyperalgesia and analgesic tolerance, effects that were not observed on chronic treatment with KGFF09. This favorable profile was further confirmed in a model of persistent inflammatory pain. In addition, we showed that KGFF09 induced less physical dependence compared with KGOP01 and KGFF03. Altogether, our data establish that combining, within a single molecule, the G-protein-biased MOPr agonism and NPFFR antagonism have beneficial effects on both acute and chronic side effects of conventional opioid analgesics. This strategy can lead to the development of novel and potent antinociceptive drugs with limited side effects on acute and chronic administration.


Subject(s)
Analgesics, Opioid/therapeutic use , Pain/drug therapy , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Animals , HEK293 Cells , Humans , Male , Mice , Motor Activity/drug effects , Pain Threshold/drug effects , Respiratory Insufficiency/chemically induced
9.
J Neurosci Methods ; 304: 92-102, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29705403

ABSTRACT

BACKGROUND: Primary microglial cultures have been used extensively to facilitate the development of therapeutic strategies for a variety of CNS disorders including neurodegeneration and neuropathic pain. However, existing techniques for culturing these cells are slow and costly. NEW METHOD: Here, we report a refined protocol based on our previously published methods described by Clark et al., which reduces in the time, reagents and the number of animals used for each culture whilst yielding high number and excellent quality microglial cells. RESULTS: Our refined protocol offers an isolation of >96% microglia from a mixed glial culture after only four days of incubation. It results in a high yield of microglia, in excess of one million cells per cortex with predominantly resting morphology and a low level of cell activation. COMPARISON WITH EXISTING METHOD(S): Compared to conventional procedures our refined protocol requires only one third of the time to prepare high quality microglial cultures, cuts the cost more than four-fold, and significantly reduces the number of animals used per culture. CONCLUSION: Our consistent, reliable, and time/cost effective microglial culture protocol is crucial for efficient in vitro screening of potential therapeutics. By dramatically reducing the culture time from 2 weeks to just 4 days and increasing the laboratory research output it has implications for the Reduction, Refinement and Replacement policies endorsed by many government funding agencies and animal research regulatory bodies.


Subject(s)
Microglia/physiology , Primary Cell Culture/economics , Primary Cell Culture/methods , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Female , Male , Rats , Rats, Wistar , Time Factors
10.
J Med Chem ; 60(22): 9407-9412, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29053268

ABSTRACT

Position 6 of the morphinan skeleton plays a key role in the µ-opioid receptor (MOR) activity in vitro and in vivo. We describe the consequence of the 6-carbonyl group deletion in N-methylmorphinan-6-ones 1-4 on ligand-MOR interaction, signaling, and antinociception. While 6-desoxo compounds 1a, 2a, and 4a show similar profiles to their 6-keto counterparts, the 6-desoxo-14-benzyloxy substituted 3a displays significantly increased MOR binding and agonist potency and a distinct binding mode compared with its analogue 3.


Subject(s)
Analgesics/pharmacology , Morphinans/pharmacology , Receptors, Opioid, mu/agonists , Analgesics/chemical synthesis , Animals , CHO Cells , Cell Membrane/physiology , Cricetulus , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/physiology , Ligands , Molecular Docking Simulation , Morphinans/chemical synthesis , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...