Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
NeuroImmune Pharm Ther ; 2(2): 187-244, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37476290

ABSTRACT

The 27th Scientific Conference of the Society on Neuroimmune Pharmacology (SNIP) in New Delhi, India, on March 15-18, 2023 is a historic summit of experts from around the world. The four day conference provides insights into the latest and most advanced science in the intersecting areas of neuroscience, immunology, pharmacology, and its translational aspects, in particular, HIV and drug abuse. Abstracts are ordered in three major groups: (1) Symposium speakers (S1-S64), (2) Investigator Posters (I1-I18), and (3) Trainee Poster (T1-T28).

2.
Neuropharmacology ; 202: 108858, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34715121

ABSTRACT

Because of increased opioid misuse, there is a need to identify new targets for minimizing opioid tolerance, and physical and psychological dependence. Previous studies showed that fibroblast growth factor 21 (FGF21) decreased alcohol and sweet preference in mice. In this study, FGF21-transgenic (FGF21-Tg) mice, expressing high FGF21 serum levels, and wildtype (WT) C57BL/6J littermates were treated with morphine and saline to determine if differences exist in their physiological and behavioral responses to opioids. FGF21-Tg mice displayed reduced preference for morphine in the conditioned place preference assay compared to WT littermates. Similarly, FGF21-Tg mice had an attenuation of the magnitude and rate of acute morphine antinociceptive tolerance development, and acute and chronic morphine physical dependence, but exhibited no change in chronic morphine antinociceptive tolerance. The ED50 values for morphine-induced antinociception in the 55 °C hot plate and the 55 °C warm-water tail withdrawal assays were similar in both strains of mice. Likewise, FGF21-Tg and WT littermates had comparable responses to morphine-induced respiratory depression. Overall, FGF21-Tg mice had a decrease in the development of acute analgesic tolerance, and the development of physical dependence, and morphine preference. FGF21 and its receptor have therapeutic potential for reducing opioid withdrawal symptoms and craving, and augmenting opioid therapeutics for acute pain patients to minimize tolerance development.


Subject(s)
Drug Tolerance/genetics , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/physiology , Morphine Dependence/genetics , Morphine/adverse effects , Nociception/drug effects , Animals , Behavior, Animal/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/genetics , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/therapy
3.
J Neuroimmune Pharmacol ; 16(4): 699-705, 2021 12.
Article in English | MEDLINE | ID: mdl-34935110

ABSTRACT

This guest commentary introduces "The Neuroimmune Pharmacology of SARS-CoV-2," a special theme issue for The Journal of Neuroimmune Pharmacology led by the Society on NeuroImmune Pharmacology. The issue builds on the Society's Virtual Workshop on COVID-19 held April 9, 2021. Top row from left: Drs. Santosh Kumar, Sowmya Yelamanchili, Pankaj Seth, Jean M. Bidlack; Bottom row from left: Drs. Gurudutt Pendyala, Sanjay Maggirwar, and Sulie L. Chang.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans
4.
J Neuroimmune Pharmacol ; 16(4): 706-717, 2021 12.
Article in English | MEDLINE | ID: mdl-34826061

ABSTRACT

In spring of 2021, the Society on NeuroImmune Pharmacology (SNIP) organized a virtual workshop on the coronavirus disease 2019 (COVID-19). The daylong event's fourth and final symposium, "Well-being and reflections," offered a glimpse at the pandemic's impact on the lives of our scientists and educators. This manuscript includes a brief summary of the symposium, a transcription of our incoming president Dr. Santosh Kumar's lecture, titled "Intervention and improved well-being of basic science researchers during the COVID-19 era: a case study," and the panel discussion that followed, "Reflection and sharing," featuring Drs. Jean M. Bidlack, Sylvia Fitting, Santhi Gorantla, Maria Cecilia G. Marcondes, Loyda M. Melendez, and Ilker K. Sariyer. The conclusion of this manuscript includes comments from SNIP's president Dr. Sulie L. Chang and our Chief Editor, Dr. Howard E. Gendelman. Drs. Sowmya Yelamanchili and Jeymohan Joseph co-chaired the symposium.


Subject(s)
COVID-19 , Humans , Pandemics , SARS-CoV-2
5.
J Neuroimmune Pharmacol ; 16(3): 519-530, 2021 09.
Article in English | MEDLINE | ID: mdl-34181181

ABSTRACT

This brief report collects the program and abstracts of the Society on NeuroImmune Pharmacology (SNIP) COVID-19 Virtual Workshop held on April 9, 2021. The workshop consisted of four symposia: Symposium 1: Molecular approaches to COVID-19 pathogenesis and underlying mechanisms; Symposium 2: Therapeutic and vaccine approaches to COVID-19; Symposium 3: Early Career Investigator talks; and Symposium 4: Diversity and Inclusion SNIP Committee (DISC) program: Well-being and reflections. The workshop also featured four special talks on COVID-19 and funding opportunities from the National Institute on Alcohol Abuse and Alcoholism (NIAAA); COVID-19 and funding opportunities from the National Institute on Drug Abuse (NIDA); opportunities from NIH for early career investigator (ECI) fellows; and neurologic and psychiatric complications of SARS-CoV-2 infection. Presenters included NIH officials, SNIP members, and non-member scientists whose abstracts were submitted and accepted for inclusion in the virtual event hosted by the University of Nebraska Medical Center via Zoom webinar. A special theme issue of SNIP's official journal, the Journal of Neuroimmune Pharmacology (JNIP), will collect select papers from the workshop along with other related manuscripts in a special theme issue titled "Neuroimmune Pharmacology of SARS-CoV-2."


Subject(s)
COVID-19 Drug Treatment , COVID-19/immunology , Education/trends , Neuroimmunomodulation/immunology , Societies, Scientific/trends , Antiviral Agents/administration & dosage , Antiviral Agents/immunology , Education/methods , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/immunology , Neuroimmunomodulation/drug effects
6.
Int J Mol Sci ; 22(1)2020 Dec 22.
Article in English | MEDLINE | ID: mdl-33374986

ABSTRACT

In previous work we evaluated an opioid glycopeptide with mixed µ/δ-opioid receptor agonism that was a congener of leu-enkephalin, MMP-2200. The glycopeptide analogue showed penetration of the blood-brain barrier (BBB) after systemic administration to rats, as well as profound central effects in models of Parkinson's disease (PD) and levodopa (L-DOPA)-induced dyskinesia (LID). In the present study, we tested the glycopeptide BBI-11008 with selective δ-opioid receptor agonism, an analogue of deltorphin, a peptide secreted from the skin of frogs (genus Phyllomedusa). We tested BBI-11008 for BBB-penetration after intraperitoneal (i.p.) injection and evaluated effects in LID rats. BBI-11008 (10 mg/kg) demonstrated good CNS-penetrance as shown by microdialysis and mass spectrometric analysis, with peak concentration levels of 150 pM in the striatum. While BBI-11008 at both 10 and 20 mg/kg produced no effect on levodopa-induced limb, axial and oral (LAO) abnormal involuntary movements (AIMs), it reduced the levodopa-induced locomotor AIMs by 50% after systemic injection. The N-methyl-D-aspartate receptor antagonist MK-801 reduced levodopa-induced LAO AIMs, but worsened PD symptoms in this model. Co-administration of MMP-2200 had been shown prior to block the MK-801-induced pro-Parkinsonian activity. Interestingly, BBI-11008 was not able to block the pro-Parkinsonian effect of MK-801 in the LID model, further indicating that a balance of mu- and delta-opioid agonism is required for this modulation. In summary, this study illustrates another example of meaningful BBB-penetration of a glycopeptide analogue of a peptide to achieve a central behavioral effect, providing additional evidence for the glycosylation technique as a method to harness therapeutic potential of peptides.


Subject(s)
Disease Models, Animal , Dyskinesia, Drug-Induced/physiopathology , Glycopeptides/pharmacology , Motor Activity/drug effects , Parkinson Disease, Secondary/physiopathology , Receptors, Opioid, delta/agonists , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/pharmacology , Animals , Corpus Striatum/metabolism , Dizocilpine Maleate/pharmacology , Dyskinesia, Drug-Induced/metabolism , Glycopeptides/administration & dosage , Glycopeptides/pharmacokinetics , Levodopa , Male , Motor Activity/physiology , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Rats, Sprague-Dawley , Receptors, Opioid, delta/metabolism
7.
Mol Pharmacol ; 98(4): 462-474, 2020 10.
Article in English | MEDLINE | ID: mdl-32958572

ABSTRACT

Opioid receptors (ORs) convert extracellular messages to signaling events by coupling to the heterotrimeric G proteins, Gα•ßγ Classic pharmacological methods, such as [35S]GTPγS binding and inhibition of cyclic AMP production, allow for general opioid characterization, but they are subject to the varying endogenous Gα proteins in a given cell type. Bioluminescence resonance energy transfer (BRET) technology offers new insight by allowing the direct observation of Gα subunit-specific effects on opioid pharmacology. Using a Venus-tagged Gßγ and nanoluciferase-tagged truncated G protein receptor kinase 3, an increase in BRET signal correlated with OR activation mediated by a specific Gα protein. The magnitude of the BRET signal was normalized to the maximum response obtained with 10 µM 2-(3,4-dichlorophenyl)-N-methyl-N-[(1R,2R)-2-pyrrolidin-1-ylcyclohexyl]acetamide (U50,488) for the kappa OR (KOR). Opioids reached equilibrium with the KOR, and concentration-response curves were generated. Although the full agonists U50,488, salvinorin A, nalfurafine, and dynorphin peptides were equally efficacious regardless of the Gα subunit present, the concentration-response curves were leftward shifted when the KOR was signaling through Gαz compared with other Gαi/o subunits. In contrast, the Gα subunit distinctly affected both the efficacy and potency of partial kappa agonists, such as the benzomorphans, and the classic mu opioid antagonists, naloxone, naltrexone, and nalmefene. For example, (-)pentazocine had EC50 values of 7.3 and 110 nM and maximal stimulation values of 79% and 35% when the KOR signaled through Gαz and Gαi1, respectively. Together, these observations suggest KOR pharmacology varies based on the specific Gα subunit coupled to the KOR. SIGNIFICANCE STATEMENT: Opioid receptors couple to various heterotrimeric Gαßγ proteins to convert extracellular cues to precise intracellular events. This paper focuses on how the various inhibitory Gα subunits influence the pharmacology of full and partial agonists at the kappa opioid receptor. Using a bioluminescent assay, the efficacy and potency of kappa opioids was determined. Opioid signaling was more potent through Gαz compared with other Gα proteins. These observations suggest that Gαz may impact opioid pharmacology and cellular physiology more than previously thought.


Subject(s)
Analgesics, Opioid/pharmacology , Bioluminescence Resonance Energy Transfer Techniques/methods , GTP-Binding Protein alpha Subunits/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Diterpenes, Clerodane/pharmacology , Dynorphins/pharmacology , HEK293 Cells , Humans , Morphinans/pharmacology , Signal Transduction/drug effects , Spiro Compounds/pharmacology
8.
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.
Nat Neurosci ; 22(11): 1782-1792, 2019 11.
Article in English | MEDLINE | ID: mdl-31636451

ABSTRACT

Microglia are the brain's resident innate immune cells and also have a role in synaptic plasticity. Microglial processes continuously survey the brain parenchyma, interact with synaptic elements and maintain tissue homeostasis. However, the mechanisms that control surveillance and its role in synaptic plasticity are poorly understood. Microglial dynamics in vivo have been primarily studied in anesthetized animals. Here we report that microglial surveillance and injury response are reduced in awake mice as compared to anesthetized mice, suggesting that arousal state modulates microglial function. Pharmacologic stimulation of ß2-adrenergic receptors recapitulated these observations and disrupted experience-dependent plasticity, and these effects required the presence of ß2-adrenergic receptors in microglia. These results indicate that microglial roles in surveillance and synaptic plasticity in the mouse brain are modulated by noradrenergic tone fluctuations between arousal states and emphasize the need to understand the effect of disruptions of adrenergic signaling in neurodevelopment and neuropathology.


Subject(s)
Microglia/physiology , Neuronal Plasticity/physiology , Norepinephrine/physiology , Visual Cortex/physiology , Animals , Benzylamines/pharmacology , CX3C Chemokine Receptor 1/genetics , Cell Movement/drug effects , Cell Movement/physiology , Circadian Rhythm/physiology , Clenbuterol/pharmacology , Dexmedetomidine/pharmacology , Dominance, Ocular , Female , Fentanyl/pharmacology , Locus Coeruleus/drug effects , Male , Mice , Mice, Transgenic , Microglia/cytology , Microglia/drug effects , Nadolol/pharmacology , Neuronal Plasticity/drug effects , Neurons/metabolism , Neurons/physiology , Norepinephrine/metabolism , Propanolamines/pharmacology , Restraint, Physical/physiology , Terbutaline/pharmacology , Wakefulness , Wounds and Injuries/physiopathology
11.
J Pharmacol Exp Ther ; 367(2): 267-281, 2018 11.
Article in English | MEDLINE | ID: mdl-30108159

ABSTRACT

A combination of buprenorphine (BUP) and samidorphan (SAM) at a 1:1 (mg/mg) fixed-ratio dose is being investigated as an adjunctive treatment of major depressive disorder (BUP/SAM, ALKS 5461). Both [3H]BUP and [3H]SAM bound to the µ-, κ-, and δ-opioid receptors (MOR, KOR, and DOR, respectively) with Kd values of 3 nM or less. [3H]BUP dissociated from the MOR more slowly than [3H]SAM did. In the [35S]GTPγS assay, BUP was a partial agonist at the MOR, KOR, and DOR. SAM was an antagonist at the MOR and a partial agonist at the KOR and DOR. The pharmacology of the combination of SAM and BUP was characterized at ratios like the molar ratios of both compounds at steady state in humans. In all assessments, SAM reduced the efficacy of BUP at the MOR without altering its potency. At the KOR, SAM had no significant effect on the activity of BUP. In bioluminescent resonance energy transfer assays, SAM, naltrexone, and naloxone were partial agonists when the MOR was coupled to the Gα oB and Gα z, and were antagonists when coupled to Gα i At the KOR, SAM was a partial agonist activating Gα oA and Gα oB and a full agonist in stimulating Gα z SAM inhibited BUP's recruitment of ß-arrestin to the MOR, suggesting an attenuation of BUP's efficacy in activating G proteins correlated with an inhibition of ß-arrestin recruitment. The collective data suggest that SAM attenuates the efficacy of BUP under all conditions tested at the MOR and DOR but had little effect on BUP activity at the KOR.


Subject(s)
Buprenorphine/pharmacology , Depressive Disorder, Major/drug therapy , Naltrexone/analogs & derivatives , Animals , CHO Cells , Cell Line , Cricetulus , Drug Combinations , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Naloxone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , beta-Arrestins/metabolism
12.
Brain Behav Immun ; 67: 152-162, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28844812

ABSTRACT

Patients receiving the cytokine immunotherapy, interferon-alpha (IFN-α) frequently present with neuropsychiatric consequences and cognitive impairments. Patients (25-80%) report symptoms of depression, including, anhedonia, irritability, fatigue and impaired motivation. Our lab has previously demonstrated treatment (170,000IU/kg sc, 3 times per week for 4weeks) of the pro-inflammatory cytokine, IFN-α, induced a depressive phenotype in rats in the forced swim test (FST). Here, we examine the biological mechanisms underlying behavioral changes induced by IFN-α, which may be reflective of mechanisms underlying inflammation associated depression. We also investigate the potential of 3-carboxamido seco-nalmefene (3CS-nalmefene), a novel opioid modulator (antagonist at mu and partial agonist at kappa and delta opioid receptors in vitro), to reverse IFN-α induced changes. In vitro radioligand receptor binding assays and the [35S] GTPγS were performed to determine the affinity of 3CS-nalmefene for the mu, kappa and delta opioid receptors. IFN-α treatment increased circulating and central markers of inflammation and hypothalamic-pituitaryadrenal (HPA) axis activity (IL-6, IL-1ß and corticosterone) while increasing immobility in the FST, impairing of object displacement learning in the object exploration task (OET), and decreasing neuronal proliferation and brain-derived neurotrophic factor (BDNF) in the hippocampus. Treatment with 3CS-nalmefene (0.3mg/kg/sc twice per day, 3 times per week for 4weeks) prevented IFN-α-induced immobility in the FST and impaired object displacement learning. In addition, 3CS-nalmefene prevented IFN-α-induced increases in inflammation and hyperactivity of the HPA-axis, the IFN-α-induced reduction in both neuronal proliferation and BDNF expression in the hippocampus. Overall, these preclinical data would support the hypothesis that opioid receptor modulation is a relevant target for treatment of depression.


Subject(s)
Antidepressive Agents/administration & dosage , Depressive Disorder/drug therapy , Naltrexone/analogs & derivatives , Narcotic Antagonists/administration & dosage , Receptors, Opioid/agonists , Animals , Anxiety/chemically induced , Anxiety/drug therapy , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/drug effects , Depression/chemically induced , Depression/drug therapy , Depressive Disorder/chemically induced , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Interferon-alpha/administration & dosage , Male , Naltrexone/administration & dosage , Neurons/drug effects , Neurons/metabolism , Rats, Wistar
13.
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
14.
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
15.
Adv Pharmacol ; 69: 387-418, 2014.
Article in English | MEDLINE | ID: mdl-24484983

ABSTRACT

Cocaine use activates the dopamine reward pathway, leading to the reinforcing effects of dopamine. There is no FDA-approved medication for treating cocaine dependence. Opioid agonists and antagonists have been approved for treating opioid and alcohol dependence. Agonists that activate the µ opioid receptor increase dopamine levels in the nucleus accumbens, while µ receptor antagonists decrease dopamine levels by blocking the effects of endogenous opioid peptides. Activation of the κ opioid receptor decreases dopamine levels and leads to dysphoria. In contrast, inhibition of the κ opioid receptor decreases dopamine levels in the nucleus accumbens. Antagonists acting at the κ receptor reduce stress-mediated behaviors and anxiety. Mixed partial µ/κ agonists have the potential of striking a balance between dopamine levels and attenuating relapse to cocaine. The pharmacological properties of mixed µ/κ opioid receptor agonists will be discussed and results from clinical and preclinical studies will be presented. Results from studies with some of the classical benzomorphans and morphinans will be presented as they lay the foundation for structure-activity relationships. Recent results with other partial opioid agonists, including buprenorphine derivatives and the mixed µ/κ peptide CJ-15,208, will be discussed. The behavioral effects of the mixed µ/κ MCL-741, an aminothiazolomorphinan, in attenuating cocaine-induced locomotor activity will be presented. While not a mixed µ/κ opioid, results obtained with GSK1521498, a µ receptor inverse agonist, will be discussed. Preclinical strategies and successes will lay the groundwork for the further development of mixed µ/κ opioid receptor agonists to treat cocaine dependence.


Subject(s)
Analgesics, Opioid/chemistry , Analgesics, Opioid/therapeutic use , Cocaine-Related Disorders/drug therapy , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Analgesics, Opioid/pharmacology , Animals , Buprenorphine/chemistry , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Cocaine-Related Disorders/metabolism , Dose-Response Relationship, Drug , Humans , Methadone/chemistry , Methadone/pharmacology , Methadone/therapeutic use , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship
16.
ACS Chem Neurosci ; 5(2): 93-9, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24393077

ABSTRACT

A series of levo- and dextromorphinan pairs have been synthesized and evaluated for their affinities to the mu, kappa, and delta opioid receptors, the N-methyl-D-aspartate (NMDA) channel, and sigma 1 and 2 receptors. It was found that levo isomers tended to have higher affinities at the opioid receptors and moderate to high affinities to the NMDA and sigma receptors, while dextro isomers tended to have lower affinities to the opioid receptors but comparatively higher affinities to the NMDA and sigma receptors. This series of compounds have interesting and complex pharmacological profiles, and merit further investigation as potential therapies for drug abuse treatment.


Subject(s)
Morphinans/pharmacology , N-Methylaspartate/drug effects , Receptors, Opioid/drug effects , Receptors, sigma/drug effects , Animals , Humans , Male , Morphinans/chemical synthesis , Rats , Rats, Sprague-Dawley
17.
J Med Chem ; 56(21): 8872-8, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24107104

ABSTRACT

Previous studies with aminothiazolomorphinans suggested that this class of opioid ligands may be useful as a potential pharmacotherapeutic to decrease drug abuse. Novel aminothiazole derivatives of cyclorphan were prepared to evaluate a series of aminothiazolomorphinans with varying pharmacological properties at the κ opioid receptor (KOR) and µ opioid receptor (MOR). This study was focused on exploring the regioisomeric analogs with the aminothiazole on the C-ring of the morphinan skeleton. Receptor binding and [(35)S]GTPγS binding assays were used to characterize the affinity and pharmacological properties of the aminothiazolomorphinans. Intracranial self-stimulation (ICSS) was used to compare the effects of a representative aminothiazolomorphinan with the morphinan mixed-KOR/MOR agonist butorphan (MCL-101) on brain-stimulation reward.


Subject(s)
Morphinans/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Thiazoles/pharmacology , Animals , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Humans , Male , Molecular Conformation , Morphinans/chemical synthesis , Morphinans/chemistry , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
18.
Behav Pharmacol ; 24(2): 144-52, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23412114

ABSTRACT

Inhibition of Gßγ-subunit signaling to phospholipase C ß3 has been shown to potentiate morphine-mediated antinociception while attenuating the development of tolerance and dependence in mice. The objective of this study was to determine the effect of Gßγ-subunit inhibition on antinociception and other pharmacological effects, such as respiratory depression, constipation, and hyperlocomotion, mediated by the µ-opioid receptor. The Gßγ-subunit inhibitor, gallein, was administered to C57BL/6J mice by intraperitoneal injection before morphine, and data were compared with mice treated with vehicle, morphine, or gallein alone. Morphine-induced antinociception was measured using the 55°C warm-water tail-withdrawal test. Pretreatment with gallein produced a dose-dependent potentiation of morphine-mediated antinociception, producing up to a 10-fold leftward shift in the morphine dose-response curve and extending the duration of antinociception induced by a single dose of morphine. Gallein pretreatment also prevented acute antinociceptive tolerance induced by morphine. In contrast, the dose-dependent respiratory depression and hyperlocomotion induced by morphine were not potentiated by gallein pretreatment. Similarly, gallein pretreatment did not potentiate morphine-conditioned place preference responses or morphine-induced constipation, as measured as a reduction in excreta. These results suggest that selectively inhibiting Gßγ-mediated signaling may selectively increase µ-opioid receptor-mediated antinociception without matching increases in adverse physiological effects.


Subject(s)
Analgesics, Opioid/agonists , Constipation/prevention & control , GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , Hyperkinesis/prevention & control , Morphine/agonists , Respiratory Insufficiency/prevention & control , Xanthenes/therapeutic use , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Analgesics, Opioid/therapeutic use , Animals , Behavior, Animal/drug effects , Constipation/chemically induced , Dose-Response Relationship, Drug , Drug Therapy, Combination , Drug Tolerance , Hyperkinesis/chemically induced , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Morphine/adverse effects , Morphine/therapeutic use , Motor Activity/drug effects , Pain Measurement , Receptors, Opioid, mu/agonists , Respiratory Insufficiency/chemically induced , Reward , Second Messenger Systems/drug effects
19.
Bioorg Med Chem Lett ; 23(7): 2128-33, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23434225

ABSTRACT

Derivatives of the lead compound N-BPE-8-CAC (1) where each CH of the biphenyl group was individually replaced by N were prepared in hopes of identifying high affinity ligands with improved aqueous solubility. Compared to 1, binding affinities of the five possible pyridinyl derivatives for the µ opioid receptor were between threefold lower to fivefold higher with the Ki of the most potent compound being 0.064 nM. Docking of 8-CAC (2) into the unliganded binding site of the mouse µ opioid receptor (pdb: 4DKL) revealed that 8-CAC and ß-FNA (from 4DKL) make nearly identical interactions with the receptor. However, for 1 and the new pyridinyl derivatives 4-8, binding is not tolerated in the 8-CAC binding mode due to the steric constraints of the large N-substituents. Either an alternative binding mode or rearrangement of the protein to accommodate these modifications may account for their high binding affinity.


Subject(s)
Biphenyl Compounds/pharmacology , Cyclopropanes/pharmacology , Pyridines/pharmacology , Receptors, Opioid/chemistry , Animals , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Cyclopropanes/chemical synthesis , Cyclopropanes/chemistry , Dose-Response Relationship, Drug , Humans , Ligands , Mice , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Receptors, Opioid/metabolism , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 22(24): 7340-4, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23142613

ABSTRACT

N-[2-(4'-methoxy[1,1'-biphenyl]-4-yl)ethyl]-8-CAC (1) is a high affinity (K(i)=0.084 nM) ligand for the µ opioid receptor and served as the lead compound for this study. Analogues of 1 were made in hopes of identifying an SAR within a series of oxygenated (distal) phenyl derivatives. A number of new analogues were made having single-digit pM affinity for the µ receptor. The most potent was the 3',4'-methylenedioxy analogue 18 (K(i)=1.6 pM).


Subject(s)
Cyclazocine/analogs & derivatives , Oxygen/chemistry , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Cyclazocine/chemical synthesis , Cyclazocine/chemistry , Cyclazocine/pharmacology , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...