Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Proc Natl Acad Sci U S A ; 120(47): e2305215120, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37972067

ABSTRACT

Transmembrane Cav2.2 (N-type) voltage-gated calcium channels are genetically and pharmacologically validated, clinically relevant pain targets. Clinical block of Cav2.2 (e.g., with Prialt/Ziconotide) or indirect modulation [e.g., with gabapentinoids such as Gabapentin (GBP)] mitigates chronic pain but is encumbered by side effects and abuse liability. The cytosolic auxiliary subunit collapsin response mediator protein 2 (CRMP2) targets Cav2.2 to the sensory neuron membrane and regulates their function via an intrinsically disordered motif. A CRMP2-derived peptide (CBD3) uncouples the Cav2.2-CRMP2 interaction to inhibit calcium influx, transmitter release, and pain. We developed and applied a molecular dynamics approach to identify the A1R2 dipeptide in CBD3 as the anchoring Cav2.2 motif and designed pharmacophore models to screen 27 million compounds on the open-access server ZincPharmer. Of 200 curated hits, 77 compounds were assessed using depolarization-evoked calcium influx in rat dorsal root ganglion neurons. Nine small molecules were tested electrophysiologically, while one (CBD3063) was also evaluated biochemically and behaviorally. CBD3063 uncoupled Cav2.2 from CRMP2, reduced membrane Cav2.2 expression and Ca2+ currents, decreased neurotransmission, reduced fiber photometry-based calcium responses in response to mechanical stimulation, and reversed neuropathic and inflammatory pain across sexes in two different species without changes in sensory, sedative, depressive, and cognitive behaviors. CBD3063 is a selective, first-in-class, CRMP2-based peptidomimetic small molecule, which allosterically regulates Cav2.2 to achieve analgesia and pain relief without negative side effect profiles. In summary, CBD3063 could potentially be a more effective alternative to GBP for pain relief.


Subject(s)
Chronic Pain , Peptidomimetics , Rats , Animals , Chronic Pain/drug therapy , Chronic Pain/metabolism , Rats, Sprague-Dawley , Peptidomimetics/pharmacology , Calcium/metabolism , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Sensory Receptor Cells/metabolism , Ganglia, Spinal/metabolism
2.
Mar Drugs ; 18(1)2019 Dec 21.
Article in English | MEDLINE | ID: mdl-31877728

ABSTRACT

Chemotherapeutic drugs are widely utilized in the treatment of human cancers. Painful chemotherapy-induced neuropathy is a common, debilitating, and dose-limiting side effect for which there is currently no effective treatment. Previous studies have demonstrated the potential utility of peptides from the marine snail from the genus Conus for the treatment of neuropathic pain. α-Conotoxin RgIA and a potent analog, RgIA4, have previously been shown to prevent the development of neuropathy resulting from the administration of oxaliplatin, a platinum-based antineoplastic drug. Here, we have examined its efficacy against paclitaxel, a chemotherapeutic drug that works by a mechanism of action distinct from that of oxaliplatin. Paclitaxel was administered at 2 mg/kg (intraperitoneally (IP)) every other day for a total of 8 mg/kg. Sprague Dawley rats that were co-administered RgIA4 at 80 µg/kg (subcutaneously (SC)) once daily, five times per week, for three weeks showed significant recovery from mechanical allodynia by day 31. Notably, the therapeutic effects reached significance 12 days after the last administration of RgIA4, which is suggestive of a rescue mechanism. These findings support the effects of RgIA4 in multiple chemotherapeutic models and the investigation of α9α10 nicotinic acetylcholine receptors (nAChRs) as a non-opioid target in the treatment of chronic pain.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Conotoxins/pharmacology , Neuralgia/drug therapy , Neuralgia/etiology , Paclitaxel/adverse effects , Animals , Humans , Hyperalgesia/drug therapy , Male , Nicotinic Antagonists/pharmacology , Rats , Rats, Sprague-Dawley
3.
Drug Dev Res ; 76(8): 432-41, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26494422

ABSTRACT

There has been recent interest in characterizing the effects of pain-like states on motivated behaviors in order to quantify how pain modulates goal-directed behavior and the persistence of that behavior. The current set of experiments assessed the effects of an incisional postoperative pain manipulation on food-maintained responding under a progressive-ratio (PR) operant schedule. Independent variables included injury state (plantar incision or anesthesia control) and reinforcer type (grain pellet or sugar pellet); dependent variables were tactile sensory thresholds and response breakpoint. Once responding stabilized on the PR schedule, separate groups of rats received a single ventral hind paw incision or anesthesia (control condition). Incision significantly reduced breakpoints in rats responding for grain, but not sugar. In rats responding for sugar, tactile hypersensitivity recovered within 24 hr, indicating a faster recovery of incision-induced tactile hypersensitivity compared to rats responding for grain, which demonstrated recovery at PD2. The NSAID analgesic, diclofenac (5.6 mg/kg) completely restored incision-depressed PR operant responding and tactile sensitivity at 3 hr following incision. The PR schedule differentiated between sucrose and grain, suggesting that relative reinforcing efficacy may be an important determinant in detecting pain-induced changes in motivated behavior.


Subject(s)
Behavior, Animal/physiology , Conditioning, Operant/physiology , Feeding Behavior/physiology , Motivation , Pain, Postoperative/physiopathology , Analgesics/pharmacology , Animals , Behavior, Animal/drug effects , Conditioning, Operant/drug effects , Diclofenac/pharmacology , Feeding Behavior/drug effects , Feeding Behavior/psychology , Hindlimb/surgery , Male , Nociception/drug effects , Nociception/physiology , Pain, Postoperative/drug therapy , Pain, Postoperative/psychology , Rats , Rats, Sprague-Dawley , Reinforcement Schedule
4.
J Pharmacol Exp Ther ; 336(3): 767-78, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21118955

ABSTRACT

We have previously reported the chemistry and antinociceptive properties of a series of glycosylated enkephalin analogs (glycopeptides) exhibiting approximately equal affinity and efficacy at δ opioid receptors (DORs) and µ opioid receptors (MORs). More detailed pharmacology of the lead glycopeptide MMP-2200 [H2N-Tyr-D-Thr-Gly-Phe-Leu-Ser-(O-ß-D-lactose)-CONH2] is presented. MMP-2200 produced dose-related antinociception in the 55°C tail-flick assay after various routes of administration. The antinociceptive effects of MMP-2200 were blocked by pretreatment with the general opioid antagonist naloxone and partially blocked by the MOR-selective antagonist ß-funaltrexamine and the DOR-selective antagonist naltrindole. The κ opioid receptor antagonist nor-binaltorphimine and the peripherally active opioid antagonist naloxone-methiodide were ineffective in blocking the antinociceptive effects of MMP-2200. At equi-antinociceptive doses, MMP-2200 produced significantly less stimulation of locomotor activity compared with morphine. Repeated administration of equivalent doses of morphine and MMP-2200 (twice daily for 3 days) produced antinociceptive tolerance (~13- and 5-fold rightward shifts, respectively). In acute and chronic physical dependence assays, naloxone precipitated a more severe withdrawal in mice receiving morphine compared with equivalent doses of the glycopeptide. Both morphine and MMP-2200 inhibited respiration and gastrointestinal transit. In summary, MMP-2200 acts as a mixed DOR/MOR agonist in vivo, which may in part account for its high antinociceptive potency after systemic administration, as well as its decreased propensity to produce locomotor stimulation, tolerance, and physical dependence in mice, compared with the MOR-selective agonist morphine. For other measures (e.g., gastrointestinal transit and respiration), the significant MOR component may not allow differentiation from morphine.


Subject(s)
Analgesics, Opioid/pharmacology , Glycopeptides/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Analgesics, Opioid/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Glycopeptides/chemistry , Humans , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Motor Activity/physiology , Pain Measurement/drug effects , Pain Measurement/methods , Receptors, Opioid, delta/physiology , Receptors, Opioid, mu/physiology
5.
Sci Rep ; 11(1): 2372, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504822

ABSTRACT

Neuronal calcium sensor 1 (NCS1) regulates a wide range of cellular functions throughout the mammalian nervous systems. Altered NCS1 expression is associated with neurodevelopmental and neurodegenerative diseases. Previous studies focused on affective and cognitive behaviors in NCS1 knockout (KO) mice, but little is known about the physiological and pathological states associated with the loss of NCS1 in the peripheral nervous system. We previously reported that NCS1 expression was reduced following paclitaxel-induced peripheral neuropathy. Here, we comprehensively investigated the phenotypes of NCS1-KO mice through a battery of behavioral tests examining both central and peripheral nervous systems. Generally, only mild differences were observed in thermal sensation and memory acquisition between NCS1-WT and -KO male mice, but not in female mice. No differences were observed in motor performance, affective behaviors, and hearing in both sexes. These results suggest that NCS1 plays a modulatory role in sensory perceptions and cognition, particularly in male mice. NCS1 has been proposed as a pharmacological target for various diseases. Therefore, the sex-specific effects of NCS1 loss may be of clinical interest. As we examined a constitutive KO model, future studies focusing on various conditional KO models will further elucidate the precise physiological significance of NCS1.


Subject(s)
Neuronal Calcium-Sensor Proteins/deficiency , Neuropeptides/deficiency , Phenotype , Psychomotor Performance , Animals , Behavior, Animal , Male , Mice , Mice, Knockout , Motor Activity , Physical Stimulation , Sensation , Temperature
6.
Pain ; 162(4): 1250-1261, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33086288

ABSTRACT

ABSTRACT: The voltage-gated sodium channel Nav1.7 is highly expressed in nociceptive afferents and is critically involved in pain signal transmission. Nav1.7 is a genetically validated pain target in humans because loss-of-function mutations cause congenital insensitivity to pain and gain-of-function mutations cause severe pain syndromes. Consequently, pharmacological inhibition has been investigated as an analgesic therapeutic strategy. We describe a small molecule Nav1.7 inhibitor, ST-2530, that is an analog of the naturally occurring sodium channel blocker saxitoxin. When evaluated against human Nav1.7 by patch-clamp electrophysiology using a protocol that favors the resting state, the Kd of ST-2530 was 25 ± 7 nM. ST-2530 exhibited greater than 500-fold selectivity over human voltage-gated sodium channel isoforms Nav1.1-Nav1.6 and Nav1.8. Although ST-2530 had lower affinity against mouse Nav1.7 (Kd = 250 ± 40 nM), potency was sufficient to assess analgesic efficacy in mouse pain models. A 3-mg/kg dose administered subcutaneously was broadly analgesic in acute pain models using noxious thermal, mechanical, and chemical stimuli. ST-2530 also reversed thermal hypersensitivity after a surgical incision on the plantar surface of the hind paw. In the spared nerve injury model of neuropathic pain, ST-2530 transiently reversed mechanical allodynia. These analgesic effects were demonstrated at doses that did not affect locomotion, motor coordination, or olfaction. Collectively, results from this study indicate that pharmacological inhibition of Nav1.7 by a small molecule agent with affinity for the resting state of the channel is sufficient to produce analgesia in a range of preclinical pain models.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Saxitoxin , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Mice , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , Protein Isoforms , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use
7.
J Pain ; 22(11): 1530-1544, 2021 11.
Article in English | MEDLINE | ID: mdl-34029686

ABSTRACT

The present experiments determined the effects of the narrow-spectrum antibiotic vancomycin on inflammatory pain-stimulated and pain-depressed behaviors in rats. Persistent inflammatory pain was modeled using dilute formalin (0.5%). Two weeks of oral vancomycin administered in drinking water attenuated Phase II formalin pain-stimulated behavior, and prevented formalin pain-depressed wheel running. Fecal microbiota transplantation produced a non-significant trend toward reversal of the vancomycin effect on pain-stimulated behavior. Vancomycin depleted Firmicutes and Bacteroidetes populations in the gut while having a partial sparing effect on Lactobacillus species and Clostridiales. The vancomycin treatment effect was associated with an altered profile in amino acid concentrations in the gut with increases in arginine, glycine, alanine, proline, valine, leucine, and decreases in tyrosine and methionine. These results indicate that vancomycin may have therapeutic effects against persistent inflammatory pain conditions that are distal to the gut. PERSPECTIVE: The narrow-spectrum antibiotic vancomycin reduces pain-related behaviors in the formalin model of inflammatory pain. These data suggest that manipulation of the gut microbiome may be one method to attenuate inflammatory pain amplitude.


Subject(s)
Amino Acids/drug effects , Anti-Bacterial Agents/pharmacology , Behavior, Animal/drug effects , Gastrointestinal Microbiome/drug effects , Inflammation/drug therapy , Motor Activity/drug effects , Pain/drug therapy , Vancomycin/pharmacology , Animals , Disease Models, Animal , Female , Inflammation/complications , Pain/etiology , Rats, Inbred F344
8.
Synapse ; 64(4): 280-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19953652

ABSTRACT

The basal (constitutive) activity of G protein-coupled receptors allows for the measurement of inverse agonist activity. Some competitive antagonists turn into inverse agonists under conditions where receptors are constitutively active. In contrast, neutral antagonists have no inverse agonist activity, and they block both agonist and inverse agonist activity. The mu-opioid receptor (MOR) demonstrates detectable constitutive activity only after a state of dependence is produced by chronic treatment with a MOR agonist. We therefore sought to identify novel MOR inverse agonists and novel neutral MOR antagonists in both untreated and agonist-treated MOR cells. CHO cells expressing the cloned human mu receptor (hMOR-CHO cells) were incubated for 20 h with medium (control) or 10 microM (2S,4aR,6aR,7R,9S,10aS,10bR)-9-(benzoyloxy)-2-(3-furanyl)dodecahydro-6a,10b-dimethyl-4,10-dioxo-2H-naphtho-[2,1-c]pyran-7-carboxylic acid methyl ester (herkinorin, HERK). HERK treatment generates a high degree of basal signaling and enhances the ability to detect inverse agonists. [(35)S]-GTP-gamma-S assays were conducted using established methods. We screened 21 MOR "antagonists" using membranes prepared from HERK-treated hMOR-CHO cells. All antagonists, including CTAP and 6beta-naltrexol, were inverse agonists. However, LTC-274 ((-)-3-cyclopropylmethyl-2,3,4,4alpha,5,6,7,7alpha-octahydro-1H-benzofuro[3,2-e]isoquinolin-9-ol)) showed the lowest efficacy as an inverse agonist, and, at concentrations less than 5 nM, had minimal effects on basal [(35)S]-GTP-gamma-S binding. Other efforts in this study identified KC-2-009 ((+)-3-((1R,5S)-2-((Z)-3-phenylallyl)-2-azabicyclo[3.3.1]nonan-5-yl)phenol hydrochloride) as an inverse agonist at untreated MOR cells. In HERK-treated cells, KC-2-009 had the highest efficacy as an inverse agonist. In summary, we identified a novel and selective MOR inverse agonist (KC-2-009) and a novel MOR antagonist (LTC-274) that shows the least inverse agonist activity among 21 MOR antagonists. LTC-274 is a promising lead compound for developing a true MOR neutral antagonist.


Subject(s)
Gene Expression Regulation/drug effects , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , Animals , CHO Cells/drug effects , CHO Cells/metabolism , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Furans/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Narcotic Antagonists/chemistry , Protein Binding , Pyrones/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Sulfur Isotopes
9.
Psychopharmacology (Berl) ; 237(4): 1195-1208, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31912192

ABSTRACT

RATIONALE AND OBJECTIVES: The present study characterized the behavioral pharmacology of a novel, mixed-action delta-selective (78:1) opioid receptor agonist, BBI-11008. This glycopeptide drug candidate was tested in assays assessing antinociception (acute, inflammatory, and neuropathic pain-like conditions) and side-effect endpoints (respiratory depression and drug self-administration). RESULTS: BBI-11008 had a 78-fold greater affinity for the delta opioid receptor than the mu receptor, and there was no binding to the kappa opioid receptor. BBI-11008 (3.2-100; 10-32 mg kg-1, i.v.) and morphine (1-10; 1-3.2 mg kg-1, i.v.) produced antinociceptive and anti-allodynic effects in assays of acute thermal nociception and complete Freund's adjuvant (CFA)-induced inflammatory pain, with BBI-11008 being less potent than morphine in both assays. BBI-11008 (1-18 mg kg-1, i.v.) had similar efficacy to gabapentin (10-56 mg kg-1, i.v.) in a spinal nerve ligation (SNL) model of neuropathic pain. In the respiration assay, with increasing %CO2 exposure, BBI-11008 produced an initial increase (32 mg kg-1, s.c.) and then decrease (56 mg kg-1, s.c.) in minute volume (MV) whereas morphine (3.2-32 mg kg-1, s.c.) produced dose-dependent decreases in MV. In the drug self-administration procedure, BBI-11008 did not maintain self-administration at any dose tested. CONCLUSIONS: These results suggest that the glycopeptide drug candidate possesses broad-spectrum antinociceptive and anti-allodynic activity across a range of pain-like conditions. Relative to morphine or fentanyl, the profile for BBI-11008 in the respiration and drug self-administration assays suggests that BBI-11008 may have less pronounced deleterious side effects. Continued assessment of this compound is warranted.


Subject(s)
Analgesics, Opioid/administration & dosage , Neuralgia/drug therapy , Pain Measurement/drug effects , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Respiratory Mechanics/drug effects , Analgesics, Opioid/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/psychology , Male , Mice , Morphine/administration & dosage , Neuralgia/metabolism , Neuralgia/psychology , Pain Measurement/psychology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Respiratory Mechanics/physiology , Self Administration
10.
J Med Chem ; 50(16): 3765-76, 2007 Aug 09.
Article in English | MEDLINE | ID: mdl-17625813

ABSTRACT

Both of the enantiomers of 5-(3-hydroxyphenyl)-N-phenylethylmorphan with C9alpha-methyl, C9-methylene, C9-keto, and C9alpha- and C9beta-hydroxy substituents were synthesized and pharmacologically evaluated. Three of the 10 compounds, (1R,5R,9S)-(-)-9-hydroxy-5-(3-hydroxyphenyl-2-phenylethyl-2-azabicyclo[3.3.1]nonane ((1R,5R,9S)-(-)-10), (1R,5S)-(+)-5-(3-hydroxyphenyl)-9-methylene-2-phenethyl-2-azabicyclo[3.3.1]nonane ((1R,5S)-(+)-14), and (1R,5S,9R)-(-)-5-(3-hydroxyphenyl)-9-methyl-2-phenethyl-2-azabicyclo[3.3.1]nonane ((1R,5S,9R)-(+)-15) had subnanomolar affinity at mu-opioid receptors (Ki = 0.19, 0.19, and 0.63 nM, respectively). The (1R,5S)-(+)-14 was found to be a mu-opioid agonist and a mu-, delta-, and kappa-antagonist in [35S]GTP-gamma-S assays and was approximately 50 times more potent than morphine in a number of acute and subchronic pain assays, including thermal and visceral models of nociception. The (1R,5R,9S)-(-)-10 compound with a C9-hydroxy substituent axially oriented to the piperidine ring (C9beta-hydroxy) was a mu-agonist about 500 times more potent than morphine. In the single-dose suppression assay, it was greater than 1000 times more potent than morphine. It is the most potent known phenylmorphan antinociceptive. The molecular structures of these compounds were energy minimized with density functional theory at the B3LYP/6-31G* level and then overlaid onto (1R,5R,9S)-(-)-10 using the heavy atoms in the morphan moiety as a common docking point. Based on modeling, the spatial arrangement of the protonated nitrogen atom and the 9beta-OH substituent in (1R,5R,9S)-(-)-10 may facilitate the alignment of a putative water chain enabling proton transfer to a nearby proton acceptor group in the mu-opioid receptor.


Subject(s)
Analgesics/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Analgesics/chemistry , Analgesics/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Haplorhini , Humans , Mice , Models, Molecular , Molecular Structure , Quantum Theory , Radioligand Assay , Receptors, Opioid, kappa/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
11.
J Med Chem ; 58(15): 5728-41, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26125201

ABSTRACT

Glycopeptides related to ß-endorphin penetrate the blood-brain barrier (BBB) of mice to produce antinociception. Two series of glycopeptides were assessed for opioid receptor binding affinity. Attempts to alter the mu-selectivity of [D-Ala(2),N-MePhe(4),Gly-ol(5)]enkephalin (DAMGO)-related glycopeptides by altering the charged residues of the amphipathic helical address were unsuccessful. A series of pan-agonists was evaluated for antinociceptive activity (55 °C tail flick) in mice. A flexible linker was required to maintain antinociceptive activity. Circular dichroism (CD) in H2O, trifluoroethanol (TFE), and SDS micelles confirmed the importance of the amphipathic helices (11s → 11sG → 11) for antinociception. The glycosylated analogues showed only nascent helices and random coil conformations in H2O. Chemical shift indices (CSI) and nuclear Overhauser effects (NOE) with 600 MHz NMR and CD confirmed helical structures in micelles, which were rationalized by molecular dynamics calculations. Antinociceptive studies with mice confirm that these glycosylated endorphin analogues are potential drug candidates that penetrate the BBB to produce potent central effects.


Subject(s)
Central Nervous System/drug effects , Glycopeptides/pharmacology , Opioid Peptides/pharmacology , Amino Acid Sequence , Animals , Circular Dichroism , Glycopeptides/chemistry , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Opioid Peptides/chemistry , Protein Conformation , Receptors, Opioid, mu/drug effects
12.
Pharmacol Biochem Behav ; 132: 49-55, 2015 05.
Article in English | MEDLINE | ID: mdl-25735493

ABSTRACT

Previous work in our laboratories provides preclinical evidence that mixed-action delta/mu receptor glycopeptides have equivalent efficacy for treating pain with reduced side effect profiles compared to widely used mu agonist analgesics such as morphine. This study evaluated the rewarding and reinforcing effects of a lead candidate, mixed-action delta/mu agonist MMP-2200, using a conditioned place preference assay as well as a drug self-administration procedure in rats. In place conditioning studies, rats underwent a 2-week conditioning protocol and were then tested for chamber preference. Rats receiving MMP-2200, at previously determined analgesic doses, could not distinguish between the drug and saline-paired chamber, whereas rats receiving the opioid agonist morphine showed a strong preference for the morphine-paired chamber. In self-administration studies, rats were trained to respond for the high efficacy mu opioid receptor agonist fentanyl on an FR5 schedule of reinforcement. Following complete dose-response determinations for fentanyl, a range of doses of MMP-2200 as well as morphine were tested. Relative to the mu agonist morphine, MMP-2200 maintained a significantly lower number of drug infusions. To begin investigating potential molecular mechanisms for the reduced side effect profile of MMP-2200, we also examined ßarrestin2 (ßarr2) recruitment and chronic MMP-2200 induced cAMP tolerance and super-activation at the human delta and mu receptors in vitro. MMP-2200 efficaciously recruited ßarr2 to both receptors, and induced cAMP tolerance and super-activation equivalent to or greater than morphine at both receptors. The in vivo findings suggest that MMP-2200 may be less reinforcing than morphine but may have some abuse potential. The reduced side effect profile cannot be explained by reduced ßarr2 recruitment or reduced cAMP tolerance and superactivation at the monomeric receptors in vitro.

13.
J Med Chem ; 57(6): 2237-46, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24576160

ABSTRACT

Glycosylated ß-endorphin analogues of various amphipathicity were studied in vitro and in vivo in mice. Opioid binding affinities of the O-linked glycopeptides (mono- or disaccharides) and unglycosylated peptide controls were measured in human receptors expressed in CHO cells. All were pan-agonists, binding to µ-, δ-, or κ-opioid receptors in the low nanomolar range (2.2-35 nM K(i)'s). The glycoside moiety was required for intravenous (i.v.) but not for intracerebroventricular (i.c.v.) activity. Circular dichroism and NMR indicated the degree of helicity in H2O, aqueous trifluoroethanol, or micelles. Glycosylation was essential for activity after i.v. administration. It was possible to manipulate the degree of helicity by the alteration of only two amino acid residues in the helical address region of the ß-endorphin analogues without destroying µ-, δ-, or κ-agonism, but the antinociceptive activity after i.v. administration could not be directly correlated to the degree of helicity in micelles.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Glycopeptides/chemistry , Glycopeptides/pharmacology , beta-Endorphin/analogs & derivatives , beta-Endorphin/pharmacology , Animals , CHO Cells , Circular Dichroism , Cricetinae , Cricetulus , Drug Design , Glycopeptides/chemical synthesis , Humans , Injections, Intravenous , Injections, Intraventricular , Magnetic Resonance Spectroscopy , Male , Mice , Micelles , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
14.
J Med Chem ; 55(19): 8350-63, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-23016952

ABSTRACT

In the search for opioid ligands with mixed functional activity, a series of 5'-(4-chlorophenyl)-4,5α-epoxypyridomorphinans possessing alkoxy or acyloxy groups at C-14 was synthesized and evaluated. In this series, the affinity and functional activity of the ligands were found to be influenced by the nature of the substituent at C-14 as well as by the substituent at N-17. Whereas the incorporation of a 3-phenylpropoxy group at C-14 on N-methylpyridomorhinan gave a dual MOR agonist/DOR agonist 17h, its incorporation on N-cyclopropylmethylpyridomorphinan gave a MOR agonist/DOR antagonist 17d. Interestingly, 17d, in contrast to 17h, did not produce tolerance or dependence effects upon prolonged treatment in cells expressing MOR and DOR. Moreover, 17d displayed greatly diminished analgesic tolerance as compared to morphine upon repeated administration, thus supporting the hypothesis that ligands with MOR agonist/DOR antagonist functional activity could emerge as novel analgesics devoid of tolerance, dependence, and related side effects.


Subject(s)
Analgesics, Opioid/chemical synthesis , Morphinans/chemical synthesis , Opioid-Related Disorders/etiology , Pyridines/chemical synthesis , Receptors, Opioid, mu/agonists , Receptors, sigma/antagonists & inhibitors , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Drug Tolerance , Humans , Male , Mice , Mice, Inbred ICR , Morphinans/adverse effects , Morphinans/pharmacology , Morphine/adverse effects , Morphine/pharmacology , Pyridines/adverse effects , Pyridines/pharmacology , Radioligand Assay , Structure-Activity Relationship
15.
Methods Enzymol ; 484: 413-43, 2010.
Article in English | MEDLINE | ID: mdl-21036244

ABSTRACT

Constitutive (basal) signaling has been described and characterized for numerous G protein coupled receptors (GPCRs). The relevance of this activity to disease, drug discovery and development, and to clinical pharmacotherapy is just beginning to emerge. Opioid receptors were the first GPCR systems for which there was definitive evidence presented for constitutive activity, with numerous studies now published on the regulation of this activity (e.g., structure/activity of the receptor as it relates to basal activity, pharmacology of ligands that act as agonists, inverse agonists and "neutral antagonists," etc.). This chapter summarizes some of the methods used to characterize constitutive activity at the mu opioid receptor (MOR) in preclinical in vitro and in vivo model systems. This includes cell-based systems that are useful for higher throughput screening of novel ligands and for studying variables that can impact basal tone in a system. In vivo assays are also described in which constitutive activity is increased in response to acute or chronic opioid agonist exposure and where withdrawal is precipitated with antagonists that may function as inverse agonists or "neutral" antagonists. The methods described have inherent advantages and disadvantages that need to be considered in any drug discovery/development program. A brief discussion of progress toward understanding the clinical implications of MOR constitutive activity in the management of opioid addiction and chronic pain is also included in this chapter.


Subject(s)
Biological Assay/methods , Receptors, Opioid, mu/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Humans
16.
Life Sci ; 86(15-16): 624-30, 2010 Apr 10.
Article in English | MEDLINE | ID: mdl-20159022

ABSTRACT

AIMS: The current study assessed the in vivo antagonist properties of nalmefene using procedures previously used to characterize the opioid antagonists naloxone, naltrexone, 6beta-naltrexol and nalbuphine. MAIN METHODS: ICR mice were used to generate antagonist dose-response curves with intraperitoneal (i.p.) nalmefene against fixed A(90) doses of morphine in models of morphine-stimulated hyperlocomotion and antinociception. Additional dose-response curves for antagonist precipitated opioid withdrawal were run in mice treated acutely (100mg/kg, s.c., -4h) or chronically (75mg pellet, s.c., -72h) with morphine. Comparisons were made between antagonist potency and degree of precipitated withdrawal. KEY FINDINGS: Nalmefene produced dose- and time-related antagonism of morphine-induced increases in locomotor activity with a calculated ID(50) (and 95% confidence interval) of 0.014 (0.007-0.027)mg/kg. Nalmefene produced rapid reversal of morphine-induced locomotor activity (5.1min for 50% reduction in morphine effect). A 0.32mg/kg dose of nalmefene produced blockade of morphine-induced antinociception in the 55 degrees C tail-flick test that lasted approximately 2h. Nalmefene was able to potently precipitate withdrawal in mice treated acutely or chronically with morphine. SIGNIFICANCE: These results demonstrate that nalmefene is similar to naloxone and naltrexone with respect to its in vivo pharmacology in mice. Specifically, nalmefene produces potent antagonism of morphine agonist effects while precipitating severe withdrawal. The compound has a slower onset and longer duration of action compared to naloxone and naltrexone. The data allows for a more complete preclinical comparison of nalmefene against other opioid antagonists including the putative opioid neutral antagonist 6beta-naltrexol.


Subject(s)
Naltrexone/analogs & derivatives , Narcotic Antagonists/pharmacology , Substance Withdrawal Syndrome , Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred ICR , Morphine/antagonists & inhibitors , Morphine/pharmacology , Motor Activity/drug effects , Naltrexone/administration & dosage , Naltrexone/pharmacology , Narcotic Antagonists/administration & dosage , Pain/drug therapy , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL