142 results on '"Selley DE"'
Search Results
2. Dissociation between the anti-allodynic effects of fingolimod (FTY720) and desensitization of S1P 1 receptor-mediated G-protein activation in a mouse model of sciatic nerve injury.
- Author
-
Pondelick AM, Moncayo LV, Donvito G, McLane VD, Gillespie JC, Hauser KF, Spiegel S, Lichtman AH, Sim-Selley LJ, and Selley DE
- Subjects
- Animals, Female, Male, Mice, Disease Models, Animal, Dose-Response Relationship, Drug, Mice, Inbred C57BL, Neuralgia drug therapy, Neuralgia metabolism, Oxadiazoles pharmacology, Receptors, Lysosphingolipid agonists, Receptors, Lysosphingolipid metabolism, Sciatic Nerve injuries, Sciatic Nerve drug effects, Sciatic Neuropathy drug therapy, Sciatic Neuropathy metabolism, Sphingosine analogs & derivatives, Sphingosine pharmacology, Sphingosine 1 Phosphate Receptor Modulators pharmacology, Fingolimod Hydrochloride pharmacology, Hyperalgesia drug therapy, Hyperalgesia metabolism, Sphingosine-1-Phosphate Receptors agonists, Sphingosine-1-Phosphate Receptors metabolism
- Abstract
Sphingosine-1-phosphate (S1P) receptor (S1PR) agonists, such as fingolimod (FTY720), alleviate nociception in preclinical pain models by either activation (agonism) or inhibition (functional antagonism) of S1PR type-1 (S1PR1). However, the dose-dependence and temporal relationship between reversal of nociception and modulation of S1PR1 signaling has not been systematically investigated. This study examined the relationship between FTY720-induced antinociception and S1PR1 adaptation using a sciatic nerve chronic constriction injury (CCI) model of neuropathic pain in male and female C57Bl/6J mice. Daily injections of FTY720 for 14 days dose-dependently reversed CCI-induced mechanical allodynia without tolerance development, and concomitantly resulted in a dose-dependent reduction of G-protein activation by the S1PR1-selective agonist SEW2871 in the lumbar spinal cord and brain. These findings indicate FTY720-induced desensitization of S1PR1 signaling coincides with its anti-allodynic effects. Consistent with this finding, a single injection of FTY720 reversed mechanical allodynia while concomitantly producing partial desensitization of S1PR1-stimulated G-protein activation in the CNS. However, mechanical allodynia returned 24-hr post injection, despite S1PR1 desensitization at that time, demonstrating a dissociation between these measures. Furthermore, CCI surgery led to elevations of sphingolipid metabolites, including S1P, which were unaffected by daily FTY720 administration, suggesting FTY720 reversed mechanical allodynia by targeting S1PR1 rather than sphingolipid metabolism. Supporting this hypothesis, acute administration of the S1PR1-selective agonist CYM-5442 mimicked the anti-allodynic effect of FTY720. In contrast, the S1PR1-selective antagonist NIBR-0213 prevented the anti-allodynic effect of FTY720, but NIBR-0213 given alone did not affect nociception. These results indicate that FTY720 alleviates CCI-induced allodynia through a mechanism distinct from functional antagonism., Competing Interests: Declaration of competing interest None., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Effects of Repeated Treatment with the Monoacylglycerol Lipase Inhibitor MJN110 on Pain-Related Depression of Nesting and Cannabinoid 1 Receptor Function in Male and Female Mice.
- Author
-
Diester CM, Balint H, Gillespie JC, Lichtman AH, Sim-Selley LJ, Selley DE, and Negus SS
- Subjects
- Animals, Female, Male, Mice, Nesting Behavior drug effects, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Depression drug therapy, Depression metabolism, Analgesics pharmacology, Analgesics therapeutic use, Benzeneacetamides pharmacology, Benzeneacetamides therapeutic use, Carbamates, Succinimides, Monoacylglycerol Lipases antagonists & inhibitors, Monoacylglycerol Lipases metabolism, Receptor, Cannabinoid, CB1 antagonists & inhibitors, Receptor, Cannabinoid, CB1 metabolism, Mice, Inbred ICR, Pain drug therapy, Pain metabolism
- Abstract
MJN110 inhibits the enzyme monoacylglycerol lipase (MAGL) to increase levels of the endocannabinoid 2-arachidonoylglycerol , an endogenous high-efficacy agonist of cannabinoid 1 and 2 receptors (CB
1/2 R). MAGL inhibitors are under consideration as candidate analgesics, and we reported previously that acute MJN110 produced partial antinociception in an assay of pain-related behavioral depression in mice. Given the need for repeated analgesic administration in many pain patients and the potential for analgesic tolerance during repeated treatment, this study examined antinociceptive effects of repeated MJN110 on pain-related behavioral depression and CB1 R-mediated G-protein function. Male and female ICR mice were treated daily for 7 days in a 2 × 2 design with (a) 1.0 mg/kg/d MJN110 or its vehicle followed by (b) intraperitoneal injection of dilute lactic acid (IP acid) or its vehicle as a visceral noxious stimulus to depress nesting behavior. After behavioral testing, G-protein activity was assessed in lumbar spinal cord (LSC) and five brain regions using an assay of CP55,940-stimulated [35 S]GTPɣS activation. As reported previously, acute MJN110 produced partial but significant relief of IP acid-induced nesting depression on day 1. After 7 days, MJN110 continued to produce significant but partial antinociception in males, while antinociceptive tolerance developed in females. Repeated MJN110 also produced modest decreases in maximum levels of CP55,940-induced [35 S]GTPɣS binding in spinal cord and most brain regions. These results indicate that repeated treatment with a relatively low antinociceptive MJN110 dose produces only partial and sex-dependent transient antinociception associated with the emergence of CB1 R desensitization in this model of IP acid-induced nesting depression. SIGNIFICANCE STATEMENT: The drug MJN110 inhibits monoacylglycerol lipase (MAGL) to increase levels of the endogenous cannabinoid 2-arachidonoylglycerol and produce potentially useful therapeutic effects including analgesia. This study used an assay of pain-related behavioral depression in mice to show that repeated MJN110 treatment produced (1) weak but sustained antinociception in male mice, (2) antinociceptive tolerance in females, and (3) modest cannabinoid-receptor desensitization that varied by region and sex. Antinociceptive tolerance may limit the utility of MJN110 for treatment of pain., (U.S. Government work not protected by U.S. copyright.)- Published
- 2024
- Full Text
- View/download PDF
4. Blocking potential metabolic sites on NAT to improve its safety profile while retaining the pharmacological profile.
- Author
-
Flammia R, Huang B, Pagare PP, M St Onge C, Abebayehu A, Gillespie JC, Mendez RE, Selley DE, Dewey WL, and Zhang Y
- Subjects
- Animals, Structure-Activity Relationship, Mice, Humans, Molecular Structure, Thiophenes chemistry, Thiophenes pharmacology, Thiophenes chemical synthesis, Thiophenes therapeutic use, Male, Dose-Response Relationship, Drug, Analgesics, Opioid pharmacology, Analgesics, Opioid chemistry, Narcotic Antagonists pharmacology, Narcotic Antagonists chemistry, Morphine pharmacology, Receptors, Opioid, mu metabolism, Receptors, Opioid, mu antagonists & inhibitors
- Abstract
The number of opioid-related overdose deaths and individuals that have suffered from opioid use disorders have significantly increased over the last 30 years. FDA approved maintenance therapies to treat opioid use disorder may successfully curb drug craving and prevent relapse but harbor adverse effects that reduce patient compliance. This has created a need for new chemical entities with improved patient experience. Previously our group reported a novel lead compound, NAT, a mu-opioid receptor antagonist that potently antagonized the antinociception of morphine and showed significant blood-brain barrier permeability. However, NAT belongs to thiophene containing compounds which are known structural alerts for potential oxidative metabolism. To overcome this, 15 NAT derivatives with various substituents at the 5'-position of the thiophene ring were designed and their structure-activity relationships were studied. These derivatives were characterized for their binding affinity, selectivity, and functional activity at the mu opioid receptor and assessed for their ability to antagonize the antinociceptive effects of morphine in vivo. Compound 12 showed retention of the basic pharmacological attributes of NAT while improving the withdrawal effects that were experienced in opioid-dependent mice. Further studies will be conducted to fully characterize compound 12 to examine whether it would serve as a new lead for opioid use disorder treatment and management., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Systematic Structure-Activity Relationship Study of Nalfurafine Analogues toward Development of Potentially Nonaddictive Pain Management Treatments.
- Author
-
St Onge CM, Pagare PP, Zheng Y, Arriaga M, Stevens DL, Mendez RE, Poklis JL, Halquist MS, Selley DE, Dewey WL, Banks ML, and Zhang Y
- Subjects
- Structure-Activity Relationship, Animals, Mice, Male, Humans, Receptors, Opioid, kappa agonists, Receptors, Opioid, kappa metabolism, Pain Management methods, Pain drug therapy, Analgesics pharmacology, Analgesics chemistry, Analgesics chemical synthesis, Analgesics therapeutic use, Spiro Compounds chemistry, Spiro Compounds pharmacology, Spiro Compounds chemical synthesis, Morphinans pharmacology, Morphinans chemistry, Morphinans chemical synthesis, Morphinans therapeutic use
- Abstract
Despite the availability of numerous pain medications, the current array of Food and Drug Administration-approved options falls short in adequately addressing pain states for numerous patients and consequently worsens the opioid crisis. Thus, it is imperative for basic research to develop novel and nonaddictive pain medications. Toward addressing this clinical goal, nalfurafine (NLF) was chosen as a lead and its structure-activity relationship (SAR) systematically studied through design, syntheses, and in vivo characterization of 24 analogues. Two analogues, 21 and 23 , showed longer durations of action than NLF in a warm-water tail immersion assay, produced in vivo effects primarily mediated by KOR and DOR, penetrated the blood-brain barrier, and did not function as reinforcers. Additionally, 21 produced fewer sedative effects than NLF. Taken together, these results aid the understanding of NLF SAR and provide insights for future endeavors in developing novel nonaddictive therapeutics to treat pain.
- Published
- 2024
- Full Text
- View/download PDF
6. Molecular Pharmacology Profiling of Phenylfentanil and Its Analogues to Understand the Putative Involvement of an Adrenergic Mechanism in Fentanyl-Induced Respiratory Depression.
- Author
-
Li M, Pagare PP, Ma H, St Onge CM, Mendez RE, Gillespie JC, Stevens DL, Dewey WL, Selley DE, and Zhang Y
- Subjects
- Receptors, Opioid, mu, Narcotic Antagonists, Analgesics, Opioid pharmacology, Fentanyl pharmacology, Adrenergic Agents
- Abstract
While there are approved therapeutics to treat opioid overdoses, the need for treatments to reverse overdoses due to ultrapotent fentanyls remains unmet. This may be due in part to an adrenergic mechanism of fentanyls in addition to their stereotypical mu-opioid receptor (MOR) effects. Herein, we report our efforts to further understanding of the functions these distinct mechanisms impart. Employing the known MOR neutral antagonist phenylfentanil as a lead, 17 analogues were designed based on the concept of isosteric replacement. To probe mechanisms of action, these analogues were pharmacologically evaluated in vitro and in vivo, while in silico modeling studies were also conducted on phenylfentanil. While it did not indicate MOR involvement in vivo, phenylfentanil yielded respiratory minute volumes similar to those caused by fentanyl. Taken together with molecular modeling studies, these results indicated that respiratory effects of fentanyls may also correlate to inhibition of both α1A- and α1B-adrenergic receptors.
- Published
- 2024
- Full Text
- View/download PDF
7. Role of efficacy as a determinant of locomotor activation by mu-opioid receptor (MOR) ligands in female and male mice. II. Effects of novel MOR-selective phenylmorphans with high-to-low MOR efficacy.
- Author
-
Santos EJ, Nassehi N, Bow EW, Chambers DR, Gutman ES, Jacobson AE, Lutz JA, Marsh SA, Rice KC, Sulima A, Selley DE, and Negus SS
- Subjects
- Animals, Female, Male, Mice, Guanosine 5'-O-(3-Thiotriphosphate), Ligands, Analgesics, Opioid pharmacology, Buprenorphine pharmacology, Receptors, Opioid, mu agonists
- Abstract
Low-efficacy mu-opioid receptor (MOR) agonists represent promising therapeutics, but existing compounds (e.g., buprenorphine, nalbuphine) span a limited range of low MOR efficacies and have poor MOR selectivity. Accordingly, new and selective low-efficacy MOR agonists are of interest. A novel set of chiral C9-substituted phenylmorphans has been reported to display improved MOR selectivity and a range of high-to-low MOR efficacies under other conditions; however, a full opioid receptor binding profile for these drugs has not been described. Additionally, studies in mice will be useful for preclinical characterization of these novel compounds, but the pharmacology of these drugs in mice has also not been examined. Accordingly, the present study characterized the binding selectivity and in vitro efficacy of these compounds using assays of opioid receptor binding and ligand-stimulated [
35 S]GTPɣS binding. Additionally, locomotor effects were evaluated as a first step for in vivo behavioral assessment in mice. The high-efficacy MOR agonist and clinically effective antidepressant tianeptine was included as a comparator. In binding studies, all phenylmorphans showed improved MOR selectivity relative to existing lower-efficacy MOR agonists. In the ligand-stimulated [35 S]GTPɣS binding assay, seven phenylmorphans had graded levels of sub-buprenorphine MOR efficacy. In locomotor studies, the compounds again showed graded efficacy with a rapid onset and ≥1 h duration of effects, evidence for MOR mediation, and minor sex differences. Tianeptine functioned as a high-efficacy MOR agonist. Overall, these in vitro and in vivo studies support the characterization of these compounds as MOR-selective ligands with graded MOR efficacy and utility for further behavioral studies in mice., (© 2023 The Authors. Pharmacology Research & Perspectives published by British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics and John Wiley & Sons Ltd.)- Published
- 2023
- Full Text
- View/download PDF
8. Structural Alterations of the "Address" Moiety of NAN Leading to the Discovery of a Novel Opioid Receptor Modulator with Reduced hERG Toxicity.
- Author
-
Ma H, Pagare PP, Li M, Neel LT, Mendez RE, Gillespie JC, Stevens DL, Dewey WL, Selley DE, and Zhang Y
- Subjects
- Humans, Ether-A-Go-Go Potassium Channels, Ligands, Receptors, Opioid, Analgesics, Opioid pharmacology
- Abstract
The search for selective opioid ligands with desired pharmacological potency and improved safety profile has always been an area of interest. Our previous effort yielded a potent opioid modulator, NAN, a 6α- N -7'-indolyl-substituted naltrexamine derivative, which exhibited promising pharmacological activities both in vitro and in vivo. However, significant human ether-a-go-go-related gene (hERG) liability limited its further development. Therefore, a systematic structural modification on NAN was conducted in order to alleviate hERG toxicity while preserving pharmacological properties, which led to the discovery of 2'-methylindolyl derivative compound 21 . Compared to NAN, compound 21 manifested overall improved pharmacological profiles. Follow-up hERG channel inhibition evaluation revealed a seven-fold decreased potency of compound 21 compared to NAN. Furthermore, several fundamental drug-like property evaluations suggested a reasonable ADME profile of 21 . Collectively, compound 21 appeared to be a promising opioid modulator for further development as a novel therapeutic agent toward opioid use disorder treatments.
- Published
- 2023
- Full Text
- View/download PDF
9. Characterization of a Potential KOR/DOR Dual Agonist with No Apparent Abuse Liability via a Complementary Structure-Activity Relationship Study on Nalfurafine Analogues.
- Author
-
Li M, Stevens DL, Arriaga M, Townsend EA, Mendez RE, Blajkevch NA, Selley DE, Banks ML, Negus SS, Dewey WL, and Zhang Y
- Subjects
- Humans, Receptors, Opioid, kappa agonists, Analgesics pharmacology, Structure-Activity Relationship, Receptors, Opioid, mu agonists, Analgesics, Opioid chemistry, Morphinans pharmacology
- Abstract
Discovery of analgesics void of abuse liability is critical to battle the opioid crisis in the United States. Among many strategies to achieve this goal, targeting more than one opioid receptor seems promising to minimize this unwanted side effect while achieving a reasonable therapeutic profile. In the process of understanding the structure-activity relationship of nalfurafine, we identified a potential analgesic agent, NMF, as a dual kappa opioid receptor/delta opioid receptor agonist with minimum abuse liability. Further characterizations, including primary in vitro ADMET studies (hERG toxicity, plasma protein binding, permeability, and hepatic metabolism), and in vivo pharmacodynamic and toxicity profiling (time course, abuse liability, tolerance, withdrawal, respiratory depression, body weight, and locomotor activity) further confirmed NMF as a promising drug candidate for future development.
- Published
- 2022
- Full Text
- View/download PDF
10. A Journey through Diastereomeric Space: The Design, Synthesis, In Vitro and In Vivo Pharmacological Activity, and Molecular Modeling of Novel Potent Diastereomeric MOR Agonists and Antagonists.
- Author
-
Chambers DR, Sulima A, Luo D, Prisinzano TE, Goldberg A, Xie B, Shi L, Paronis CA, Bergman J, Nassehi N, Selley DE, Imler GH, Jacobson AE, and Rice KC
- Subjects
- Animals, CHO Cells, Colforsin, Cricetinae, Ligands, Mice, Morphine pharmacology, Receptors, Opioid metabolism, Receptors, Opioid, delta metabolism, Receptors, Opioid, mu metabolism, Naltrexone, Respiratory Insufficiency
- Abstract
Four sets of diastereomeric C9-alkenyl 5-phenylmorphans, varying in the length of the C9-alkenyl chain, were designed to examine the effect of these spatially distinct ligands on opioid receptors. Functional activity was obtained by forskolin-induced cAMP accumulation assays and several compounds were examined in the [
35 S]GTPgS assay and in an assay for respiratory depression. In each of the four sets, similarities and differences were observed dependent on the length of their C9-alkenyl chain and, most importantly, their stereochemistry. Three MOR antagonists were found to be as or more potent than naltrexone and, unlike naltrexone, none had MOR, KOR, or DOR agonist activity. Several potent MOR full agonists were obtained, and, of particular interest partial agonists were found that exhibited less respiratory depression than that caused by morphine. The effect of stereochemistry and the length of the C9-alkenyl chain was also explored using molecular modeling. The MOR antagonists were found to interact with the inactive (4DKL) MOR crystal structures and agonists were found to interact with the active (6DDF) MOR crystal structures. The comparison of their binding modes at the mouse MOR was used to gain insight into the structural basis for their stereochemically induced pharmacological differences.- Published
- 2022
- Full Text
- View/download PDF
11. Rational Design, Chemical Syntheses, and Biological Evaluations of Peripherally Selective Mu Opioid Receptor Ligands as Potential Opioid Induced Constipation Treatment.
- Author
-
Huang B, Li M, Klongkumnuankarn P, Mendez RE, Gillespie JC, Stevens DL, Dewey WL, Selley DE, and Zhang Y
- Subjects
- Analgesics, Opioid adverse effects, Constipation chemically induced, Constipation drug therapy, Humans, Ligands, Naltrexone pharmacology, Naltrexone therapeutic use, Narcotic Antagonists pharmacology, Narcotic Antagonists therapeutic use, Receptors, Opioid, mu, Opioid-Induced Constipation
- Abstract
Opioid-induced constipation (OIC) is a common adverse effect of opioid analgesics. Peripherally acting μ opioid receptor antagonists (PAMORAs) can be applied in the treatment of OIC without compromising the analgesic effects. NAP, a 6β-N-4-pyridyl-substituted naltrexamine derivative, was previously identified as a potent and selective MOR antagonist mainly acting peripherally but with some CNS effects. Herein, we introduced a highly polar aromatic moiety, for example, a pyrazolyl or imidazolyl ring to decrease CNS MPO scores in order to reduce passive BBB permeability. Four compounds 2 , 5 , 17 , and 19 , when administered orally, were able to increase intestinal motility during morphine-induced constipation in the carmine red dye assays. Among them, compound 19 (p.o.) improved GI tract motility by 75% while orally administered NAP and methylnaltrexone showed no significant effects at the same dose. Thus, this compound seemed a promising agent to be further developed as an oral treatment for OIC.
- Published
- 2022
- Full Text
- View/download PDF
12. Design, Synthesis, and Biological Evaluation of NAP Isosteres: A Switch from Peripheral to Central Nervous System Acting Mu-Opioid Receptor Antagonists.
- Author
-
Pagare PP, Li M, Zheng Y, Kulkarni AS, Obeng S, Huang B, Ruiz C, Gillespie JC, Mendez RE, Stevens DL, Poklis JL, Halquist MS, Dewey WL, Selley DE, and Zhang Y
- Subjects
- Analgesics, Opioid chemistry, Central Nervous System, Humans, Naloxone, Narcotic Antagonists pharmacology, Narcotic Antagonists therapeutic use, Receptors, Opioid, mu, Morphinans chemistry, Opioid-Related Disorders drug therapy
- Abstract
The μ opioid receptor (MOR) has been an intrinsic target to develop treatment of opioid use disorders (OUD). Herein, we report our efforts on developing centrally acting MOR antagonists by structural modifications of 17-cyclopropylmethyl-3,14-dihydroxy-4,5α-epoxy-6β-[(4'-pyridyl) carboxamido] morphinan (NAP), a peripherally acting MOR-selective antagonist. An isosteric replacement concept was applied and incorporated with physiochemical property predictions in the molecular design. Three analogs, namely, 25 , 26 , and 31 , were identified as potent MOR antagonists in vivo with significantly fewer withdrawal symptoms than naloxone observed at similar doses. Furthermore, brain and plasma drug distribution studies supported the outcomes of our design strategy on these compounds. Taken together, our isosteric replacement of pyridine with pyrrole, furan, and thiophene provided insights into the structure-activity relationships of NAP and aided the understanding of physicochemical requirements of potential CNS acting opioids. These efforts resulted in potent, centrally efficacious MOR antagonists that may be pursued as leads to treat OUD.
- Published
- 2022
- Full Text
- View/download PDF
13. Novel bivalent ligands carrying potential antinociceptive effects by targeting putative mu opioid receptor and chemokine receptor CXCR4 heterodimers.
- Author
-
Ma H, Li M, Pagare PP, Wang H, Nassehi N, Santos EJ, Stevens Negus S, Selley DE, and Zhang Y
- Subjects
- Animals, Ligands, Mice, Models, Molecular, Signal Transduction, Analgesics pharmacology, Analgesics therapeutic use, Receptors, Opioid, mu
- Abstract
The functional interactions between opioid and chemokine receptors have been implicated in the pathological process of chronic pain. Mounting studies have indicated the possibility that a MOR-CXCR4 heterodimer may be involved in nociception and related pharmacologic effects. Herein we have synthesized a series of bivalent ligands containing both MOR agonist and CXCR4 antagonist pharmacophores with an aim to investigate the functional interactions between these two receptors. In vitro studies demonstrated reasonable recognition of designed ligands at both respective receptors. Further antinociceptive testing in mice revealed compound 1a to be the most promising member of this series. Additional molecular modeling studies corroborated the findings observed. Taken together, we identified the first bivalent ligand 1a showing promising antinociceptive effect by targeting putative MOR-CXCR4 heterodimers, which may serve as a novel chemical probe to further develop more potent bivalent ligands with potential application in analgesic therapies for chronic pain management., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
14. Structure-Based Design and Development of Chemical Probes Targeting Putative MOR-CCR5 Heterodimers to Inhibit Opioid Exacerbated HIV-1 Infectivity.
- Author
-
Huang B, Wang H, Zheng Y, Li M, Kang G, Barreto-de-Souza V, Nassehi N, Knapp PE, Selley DE, Hauser KF, and Zhang Y
- Subjects
- Analgesics, Opioid pharmacology, Anti-HIV Agents chemistry, Anti-HIV Agents metabolism, Anti-HIV Agents pharmacology, Binding Sites, Dimerization, HIV-1 drug effects, HIV-1 physiology, Humans, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear metabolism, Leukocytes, Mononuclear virology, Maraviroc chemistry, Molecular Docking Simulation, Molecular Dynamics Simulation, Naltrexone chemistry, Phytohemagglutinins pharmacology, Protein Binding, Receptors, CCR5 chemistry, Receptors, Opioid, mu chemistry, Virus Internalization drug effects, Drug Design, Ligands, Receptors, CCR5 metabolism, Receptors, Opioid, mu metabolism
- Abstract
Crystal structures of ligand-bound G-protein-coupled receptors provide tangible templates for rationally designing molecular probes. Herein, we report the structure-based design, chemical synthesis, and biological investigations of bivalent ligands targeting putative mu opioid receptor C-C motif chemokine ligand 5 (MOR-CCR5) heterodimers. The bivalent ligand VZMC013 possessed nanomolar level binding affinities for both the MOR and CCR5, inhibited CCL5-stimulated calcium mobilization, and remarkably improved anti-HIV-1
BaL activity over previously reported bivalent ligands. VZMC013 inhibited viral infection in TZM-bl cells coexpressing CCR5 and MOR to a greater degree than cells expressing CCR5 alone. Furthermore, VZMC013 blocked human immunodeficiency virus (HIV)-1 entry in peripheral blood mononuclear cells (PBMC) cells in a concentration-dependent manner and inhibited opioid-accelerated HIV-1 entry more effectively in phytohemagglutinin-stimulated PBMC cells than in the absence of opioids. A three-dimensional molecular model of VZMC013 binding to the MOR-CCR5 heterodimer complex is constructed to elucidate its mechanism of action. VZMC013 is a potent chemical probe targeting MOR-CCR5 heterodimers and may serve as a pharmacological agent to inhibit opioid-exacerbated HIV-1 entry.- Published
- 2021
- Full Text
- View/download PDF
15. Exploring naltrexamine derivatives featuring azaindole moiety via nitrogen-walk approach to investigate their in vitro pharmacological profiles at the mu opioid receptor.
- Author
-
Ma H, Wang H, Gillespie JC, Mendez RE, Selley DE, and Zhang Y
- Subjects
- Binding Sites, Humans, Ligands, Models, Molecular, Molecular Docking Simulation, Naltrexone chemical synthesis, Naltrexone pharmacology, Opioid-Related Disorders drug therapy, Protein Conformation, Naltrexone analogs & derivatives, Nitrogen chemistry, Receptors, Opioid, mu drug effects
- Abstract
In the present work, we reported the application of a nitrogen-walk approach on developing a series of novel opioid ligands containing an azaindole moiety at the C6-position of the epoxymorphinan skeleton. In vitro study results showed that introducing a nitrogen atom around the indole moiety not only retained excellent binding affinity, but also led to significant functional switch at the mu opioid receptor (MOR). Further computational investigations provided corroborative evidence and plausible explanations of the results of the in vitro studies. Overall, our current work implemented a series of novel MOR ligands with high binding affinity and considerably low efficacy, which may shed light on rational design of low efficacy MOR ligands for opioid use disorder therapeutics., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
16. Verifying the role of 3-hydroxy of 17-cyclopropylmethyl-4,5α-epoxy-3,14β-dihydroxy-6β-[(4'-pyridyl) carboxamido]morphinan derivatives via their binding affinity and selectivity profiles on opioid receptors.
- Author
-
Huang B, Gunta R, Wang H, Li M, Cao D, Mendez RE, Gillespie JC, Chen C, Huang LM, Liu-Chen LY, Selley DE, and Zhang Y
- Subjects
- Models, Molecular, Molecular Docking Simulation, Molecular Structure, Protein Binding, Receptors, Opioid chemistry, Morphinans chemistry, Morphinans pharmacology, Receptors, Opioid metabolism
- Abstract
In the present study, the role of 3-hydroxy group of a series of epoxymorphinan derivatives in their binding affinity and selectivity profiles toward the opioid receptors (ORs) has been investigated. It was found that the 3-hydroxy group was crucial for the binding affinity of these derivatives for all three ORs due to the fact that all the analogues 1a-e exhibited significantly higher binding affinities compared to their counterpart 3-dehydroxy ones 6a-e. Meanwhile most compounds carrying the 3-hydroxy group possessed similar selectivity profiles for the kappa opioid receptor over the mu opioid receptor as their corresponding 3-dehydroxy derivatives. [
35 S]-GTPγS functional assay results indicated that the 3-hydroxy group of these epoxymorphinan derivatives was important for maintaining their potency on the ORs with various effects. Further molecular modeling studies helped comprehend the remarkably different binding affinity and functional profiles between compound 1c (NCP) and its 3-dehydroxy analogue 6c., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
17. Exploring the putative mechanism of allosteric modulations by mixed-action kappa/mu opioid receptor bitopic modulators.
- Author
-
Wang H, Cao D, Gillespie JC, Mendez RE, Selley DE, Liu-Chen LY, and Zhang Y
- Subjects
- Allosteric Regulation, Allosteric Site, Binding Sites, Humans, Ligands, Molecular Docking Simulation, Morphinans chemistry, Morphinans metabolism, Naltrexone analogs & derivatives, Naltrexone chemistry, Naltrexone metabolism, Protein Binding, Receptors, Opioid, kappa chemistry, Receptors, Opioid, mu chemistry, Spiro Compounds chemistry, Spiro Compounds metabolism, Thermodynamics, Receptors, Opioid, kappa metabolism, Receptors, Opioid, mu metabolism
- Abstract
The modulation and selectivity mechanisms of seven mixed-action kappa opioid receptor (KOR)/mu opioid receptor (MOR) bitopic modulators were explored. Molecular modeling results indicated that the 'message' moiety of seven bitopic modulators shared the same binding mode with the orthosteric site of the KOR and MOR, whereas the 'address' moiety bound with different subdomains of the allosteric site of the KOR and MOR. The 'address' moiety of seven bitopic modulators bound to different subdomains of the allosteric site of the KOR and MOR may exhibit distinguishable allosteric modulations to the binding affinity and/or efficacy of the 'message' moiety. Moreover, the 3-hydroxy group on the phenolic moiety of the seven bitopic modulators induced selectivity to the KOR over the MOR.
- Published
- 2021
- Full Text
- View/download PDF
18. Manipulating Pharmacodynamic Efficacy with Agonist + Antagonist Mixtures: In Vitro and In Vivo Studies with Opioids and Cannabinoids.
- Author
-
Selley DE, Banks ML, Diester CM, Jali AM, Legakis LP, Santos EJ, and Negus SS
- Subjects
- Animals, CHO Cells, Cricetulus, Dose-Response Relationship, Drug, Drug Interactions, Male, Mice, Receptor, Cannabinoid, CB1 agonists, Receptor, Cannabinoid, CB1 antagonists & inhibitors, Receptors, Opioid, mu agonists, Receptors, Opioid, mu antagonists & inhibitors, Analgesics, Opioid pharmacology, Cannabinoids pharmacology
- Abstract
Pharmacodynamic efficacy of drugs to activate their receptors is a key determinant of drug effects, and intermediate-efficacy agonists are often useful clinically because they retain sufficient efficacy to produce therapeutically desirable effects while minimizing undesirable effects. Molecular mechanisms of efficacy are not well understood, so rational drug design to control efficacy is not yet possible; however, receptor theory predicts that fixed-proportion mixtures of an agonist and antagonist for a given receptor can be adjusted to precisely control net efficacy of the mixture in activating that receptor. Moreover, the agonist proportion required to produce different effects provides a quantitative scale for comparing efficacy requirements across those effects. To test this hypothesis, the present study evaluated effectiveness of fixed-proportion agonist/antagonist mixtures to produce in vitro and in vivo effects mediated by μ -opioid receptors (MOR) and cannabinoid type 1 receptors (CB
1 R). Mixtures of 1) the MOR agonist fentanyl and antagonist naltrexone and 2) the CB1 R agonist CP55,940 and antagonist/inverse agonist rimonabant were evaluated in an in vitro assay of ligand-stimulated guanosine 5'- O -(3-[35 S]thio)triphosphate binding and an in vivo assay of thermal nociception in mice. For both agonist/antagonist pairs in both assays, increasing agonist proportions produced graded increases in maximal mixture effects, and lower agonist proportions were sufficient to produce in vivo than in vitro effects. These findings support the utility of agonist-antagonist mixtures as a strategy to control net efficacy of receptor activation and to quantify and compare efficacy requirements across a range of in vitro and in vivo endpoints. SIGNIFICANCE STATEMENT: Manipulation of agonist proportion in agonist/antagonist mixtures governs net mixture efficacy at the target receptor. Parameters of agonist/antagonist mixture effects can provide a quantitative metric for comparison of efficacy requirements across a wide range of conditions., Competing Interests: No author has an actual or perceived conflict of interest with the contents of this article., (Copyright © 2021 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2021
- Full Text
- View/download PDF
19. Bivalent Ligand Aiming Putative Mu Opioid Receptor and Chemokine Receptor CXCR4 Dimers in Opioid Enhanced HIV-1 Entry.
- Author
-
Ma H, Wang H, Li M, Barreto-de-Souza V, Reinecke BA, Gunta R, Zheng Y, Kang G, Nassehi N, Zhang H, An J, Selley DE, Hauser KF, and Zhang Y
- Abstract
A bivalent compound 1a featuring both a mu opioid receptor (MOR) and a CXCR4 antagonist pharmacophore (naltrexone and IT1t) was designed and synthesized. Further binding and functional studies demonstrated 1a acting as a MOR and a CXCR4 dual antagonist with reasonable binding affinities at both receptors. Furthermore, compound 1a seemed more effective than a combination of IT1t and naltrexone in inhibiting HIV entry at the presence of morphine. Additional molecular modeling results suggested that 1a may bind with the putative MOR-CXCR4 heterodimer to induce its anti-HIV activity. Collectively, bivalent ligand 1a may serve as a promising lead to develop chemical probes targeting the putative MOR-CXCR4 heterodimer in comprehending opioid exacerbated HIV-1 invasion., Competing Interests: The authors declare no competing financial interest.
- Published
- 2020
- Full Text
- View/download PDF
20. Kappa opioid receptors mediate an initial aversive component of paclitaxel-induced neuropathy.
- Author
-
Meade JA, Alkhlaif Y, Contreras KM, Obeng S, Toma W, Sim-Selley LJ, Selley DE, and Damaj MI
- Subjects
- Amygdala drug effects, Amygdala metabolism, Animals, Avoidance Learning physiology, Dose-Response Relationship, Drug, Guanosine 5'-O-(3-Thiotriphosphate) metabolism, Male, Mice, Mice, Inbred C57BL, Nucleus Accumbens drug effects, Nucleus Accumbens metabolism, Antineoplastic Agents, Phytogenic toxicity, Avoidance Learning drug effects, Paclitaxel toxicity, Peripheral Nervous System Diseases chemically induced, Peripheral Nervous System Diseases metabolism, Receptors, Opioid, kappa metabolism
- Abstract
Rationale: Cancer patients receiving the antineoplastic drug paclitaxel report higher incidences and longer duration of treatment-resistant depression than patients receiving other classes of chemotherapeutics. Rodents treated with paclitaxel exhibit a suite of changes in affect-like behaviors. Further, paclitaxel causes chemotherapy-induced peripheral neuropathy (CIPN) in humans and rodents. Kappa opioid receptors (KOR) have a well-established role in depression and neuropathy. The contributions of KOR signaling to paclitaxel-induced aversive-like state and CIPN in rodents remain to be explored., Objectives: We aimed to investigate whether dysregulation of the KOR/dynorphin system is associated with paclitaxel-mediated pain-like behavior and depression-like behavior., Methods: Cancer-free male C57BL/6J mice were treated with four injections of vehicle or paclitaxel (32 mg/kg cumulative). The effects of the selective KOR antagonist norbinaltorphimine (norBNI) on paclitaxel-induced sucrose preference deficits and mechanical hypersensitivity were measured. Prodynorphin mRNA and receptor-mediated G protein activation were measured at two time points following the last paclitaxel injection using quantitative real-time polymerase chain reaction and agonist-stimulated [
35 S]guanosine-5'-O'-(γ-thio)-triphosphate ([35 S]GTPγS) binding, respectively, in the nucleus accumbens (NAc), caudate-putamen, amygdala, and spinal cord., Results: Paclitaxel produced a norBNI-reversible sucrose preference deficit, whereas mechanical hypersensitivity was not reversed by norBNI. Paclitaxel treatment increased the levels of mRNA for prodynorphin, a precursor for endogenous KOR agonists, in the NAc. Paclitaxel also had time-dependent effects on KOR-mediated G protein activation in the NAc., Conclusions: These results suggest that KOR signaling mediates an initial aversive component of paclitaxel, but not necessarily paclitaxel-induced mechanical hypersensitivity.- Published
- 2020
- Full Text
- View/download PDF
21. Attenuated dopamine receptor signaling in nucleus accumbens core in a rat model of chemically-induced neuropathy.
- Author
-
Selley DE, Lazenka MF, Sim-Selley LJ, Secor McVoy JR, Potter DN, Chartoff EH, Carlezon WA Jr, and Negus SS
- Subjects
- Animals, Conditioning, Operant drug effects, Conditioning, Operant physiology, Disease Models, Animal, Dopamine Agonists pharmacology, Dopamine Antagonists pharmacology, Dose-Response Relationship, Drug, Male, Neuralgia chemically induced, Nucleus Accumbens drug effects, Rats, Rats, Sprague-Dawley, Receptors, Dopamine D1 agonists, Receptors, Dopamine D1 antagonists & inhibitors, Receptors, Dopamine D2 agonists, Signal Transduction drug effects, Formaldehyde toxicity, Neuralgia metabolism, Nucleus Accumbens metabolism, Receptors, Dopamine D1 metabolism, Receptors, Dopamine D2 metabolism, Signal Transduction physiology
- Abstract
Neuropathy is major source of chronic pain that can be caused by mechanically or chemically induced nerve injury. Intraplantar formalin injection produces local necrosis over a two-week period and has been used to model neuropathy in rats. To determine whether neuropathy alters dopamine (DA) receptor responsiveness in mesolimbic brain regions, we examined dopamine D
1 -like and D2 -like receptor (D1/2 R) signaling and expression in male rats 14 days after bilateral intraplantar formalin injections into both rear paws. D2 R-mediated G-protein activation and expression of the D2 R long, but not short, isoform were reduced in nucleus accumbens (NAc) core, but not in NAc shell, caudate-putamen or ventral tegmental area of formalin- compared to saline-treated rats. In addition, D1 R-stimulated adenylyl cyclase activity was also reduced in NAc core, but not in NAc shell or prefrontal cortex, of formalin-treated rats, whereas D1 R expression was unaffected. Other proteins involved in dopamine neurotransmission, including dopamine uptake transporter and tyrosine hydroxylase, were unaffected by formalin treatment. In behavioral tests, the potency of a D2 R agonist to suppress intracranial self-stimulation (ICSS) was decreased in formalin-treated rats, whereas D1 R agonist effects were not altered. The combination of reduced D2 R expression and signaling in NAc core with reduced suppression of ICSS responding by a D2 R agonist suggest a reduction in D2 autoreceptor function. Altogether, these results indicate that intraplantar formalin produces attenuation of highly specific DA receptor signaling processes in NAc core of male rats and suggest the development of a neuropathy-induced allostatic state in both pre- and post-synaptic DA receptor function., (Copyright © 2020. Published by Elsevier Ltd.)- Published
- 2020
- Full Text
- View/download PDF
22. Application of Bivalent Bioisostere Concept on Design and Discovery of Potent Opioid Receptor Modulators.
- Author
-
Ma H, Obeng S, Wang H, Zheng Y, Li M, Jali AM, Stevens DL, Dewey WL, Selley DE, and Zhang Y
- Subjects
- Animals, Calcium metabolism, Cyclic AMP metabolism, Ligands, Male, Mice, Opioid-Related Disorders drug therapy, Radioligand Assay, Signal Transduction, Structure-Activity Relationship, Drug Design, Drug Discovery, Morphinans chemistry, Morphinans pharmacology, Narcotic Antagonists chemistry, Narcotic Antagonists pharmacology, Receptors, Opioid, mu antagonists & inhibitors
- Abstract
Here, we described the structural modification of previously identified μ opioid receptor (MOR) antagonist NAN, a 6α- N -7'-indolyl substituted naltrexamine derivative, and its 6β- N -2'-indolyl substituted analogue INTA by adopting the concept of "bivalent bioisostere". Three newly prepared opioid ligands, 25 (NBF), 31 , and 38 , were identified as potent MOR antagonists both in vitro and in vivo. Moreover, these three compounds significantly antagonized DAMGO-induced intracellular calcium flux and displayed varying degrees of inhibition on cAMP production. Furthermore, NBF produced much less significant withdrawal effects than naloxone in morphine-pelleted mice. Molecular modeling studies revealed that these bivalent bioisosteres may adopt similar binding modes in the MOR and the "address" portions of them may have negative or positive allosteric modulation effects on the function of their "message" portions compared with NAN and INTA. Collectively, our successful application of the "bivalent bioisostere concept" identified a promising lead to develop novel therapeutic agents toward opioid use disorder treatments.
- Published
- 2019
- Full Text
- View/download PDF
23. Design and synthesis of a bivalent probe targeting the putative mu opioid receptor and chemokine receptor CXCR4 heterodimer.
- Author
-
Reinecke BA, Kang G, Zheng Y, Obeng S, Zhang H, Selley DE, An J, and Zhang Y
- Abstract
Opioid abuse and HIV/AIDS have been defined as synergistic epidemics. Opioids can accelerate HIV replication in the immune system by up-regulating the expression of HIV co-receptor CXCR4. Several hypotheses have been suggested as the mechanism of CXCR4 modulation by opioids through their activation on the mu opioid receptor (MOR). One hypothesis is the putative heterodimerization of the MOR and CXCR4 as a mechanism of cross-talk and subsequent exacerbation of HIV replication. Bivalent chemical probes can be powerful molecular tools to characterize protein-protein interactions, and modulate the function related to such interactions. Herein we report the design and synthesis of a novel bivalent probe to explore the putative MOR-CXCR4 dimerization and its potential pharmacological role in enhancing HIV progression. The developed bivalent probe was designed with two distinct pharmacophores linked through a spacer. One pharmacophore (naltrexone) will interact with the MOR and the other (IT1t) with the CXCR4. The overall synthetic routes to prepare the bivalent probe and its corresponding monovalent controls were comprised of 18-22 steps with acceptable yields. Preliminary biological evaluation showed that the bivalent probe preserved binding affinity and functional activity at both respective receptors, supporting the initial molecular design., (This journal is © The Royal Society of Chemistry 2020.)
- Published
- 2019
- Full Text
- View/download PDF
24. Pharmacological characterization of 17-cyclopropylmethyl-3,14-dihydroxy-4,5-epoxy-6-[(3'-fluoro-4'-pyridyl)acetamido]morphinan (NFP) as a dual selective MOR/KOR ligand with potential applications in treating opioid use disorder.
- Author
-
Zheng Y, Obeng S, Reinecke BA, Chen C, Phansalkar PS, Walentiny DM, Gerk PM, Liu-Chen LY, Selley DE, Beardsley PM, and Zhang Y
- Subjects
- Analgesics, Opioid pharmacology, Animals, Biological Transport, CHO Cells, Caco-2 Cells, Calcium metabolism, Cell Line, Tumor, Cricetulus, Enkephalin, Ala(2)-MePhe(4)-Gly(5)- pharmacology, Humans, Ligands, Male, Mice, Inbred C57BL, Narcotic Antagonists pharmacology, Receptors, Opioid, kappa agonists, Receptors, Opioid, mu agonists, Receptors, Opioid, mu genetics, Receptors, Opioid, mu metabolism, Morphinans pharmacology, Opioid-Related Disorders drug therapy, Receptors, Opioid, kappa antagonists & inhibitors, Receptors, Opioid, mu antagonists & inhibitors
- Abstract
For thousands of years opioids have been the first-line treatment option for pain management. However, the tolerance and addiction potential of opioids limit their applications in clinic. NFP, a MOR/KOR dual-selective opioid antagonist, was identified as a ligand that significantly antagonized the antinociceptive effects of morphine with lesser withdrawal effects than naloxone at similar doses. To validate the potential application of NFP in opioid addiction treatment, a series of in vitro and in vivo assays were conducted to further characterize its pharmacological profile. In calcium mobilization assays and MOR internalization studies, NFP showed the apparent capacity to antagonize DAMGO-induced calcium flux and etorphine-induced MOR internalization. In contrast to the opioid agonists DAMGO and morphine, cells pretreated with NFP did not show apparent desensitization and down regulation of the MOR. Though in vitro bidirectional transport studies showed that NFP might be a P-gp substrate, in warm-water tail-withdrawal assays it was able to antagonize the antinociceptive effects of morphine indicating its potential central nervous system activity. Overall these results suggest that NFP is a promising dual selective opioid antagonist that may have the potential to be used therapeutically in opioid use disorder treatment., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
25. Gnal haploinsufficiency causes genomic instability and increased sensitivity to haloperidol.
- Author
-
Khan MM, Xiao J, Hollingsworth TJ, Patel D, Selley DE, Ring TL, and LeDoux MS
- Subjects
- Animals, Brain drug effects, Brain pathology, Female, Haploinsufficiency, Male, Mice, Tardive Dyskinesia genetics, Dopamine Antagonists pharmacology, GTP-Binding Protein alpha Subunits genetics, Genomic Instability drug effects, Haloperidol pharmacology
- Abstract
GNAL encodes guanine nucleotide-binding protein subunit Gα(olf) which plays a key role in striatal medium spiny neuron (MSN)-dopamine signaling. GNAL loss-of-function mutations are causally-associated with isolated dystonia, a movement disorder characterized by involuntary muscle contractions leading to abnormal postures. Dopamine D2 receptor (D2R) blockers such as haloperidol are mainstays in the treatment of psychosis but may contribute to the development of secondary acute and tardive dystonia. Administration of haloperidol promotes cAMP-dependent signaling in D2R-expressing indirect pathway MSNs. At present, little is known about the cellular relationships among isolated, acute, and tardive dystonia. Herein, we report the effects of acute D2R blockade on motor behavior, DNA repair, cAMP-mediated histone H3 phosphorylation (Ser10), and cell death in Gnal
+/- mice and their isogenic Gnal+/+ littermates. In comparison to Gnal+/+ littermates, Gnal+/- mice exhibited increased catalepsy responses, persistent DNA breaks, decreased cAMP-dependent histone H3 phosphorylation (Ser10), and increased cell death in response to haloperidol. In striatum, aged Gnal+/- mice exhibited increased global DNA methylation, increased euchromatin, and dendritic structural abnormalities. Our results provide evidence that Gα(olf) deficiency intensifies the effects of D2R antagonism and suggests that loss-of-function variants in GNAL may increase risk for movement disorders associated with D2R blockers. We hypothesize that the effects of Gα(olf) dysfunction and/or long-term D2R antagonism may lead to epigenetic silencing, transcriptional dysregulation, and, ultimately, cellular senescence and/or apoptosis in human brain., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
26. Characterization of 17-Cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(indole-7-carboxamido)morphinan (NAN) as a Novel Opioid Receptor Modulator for Opioid Use Disorder Treatment.
- Author
-
Obeng S, Jali A, Zheng Y, Wang H, Schwienteck KL, Chen C, Stevens DL, Akbarali HI, Dewey WL, Banks ML, Liu-Chen LY, Selley DE, and Zhang Y
- Subjects
- Analgesics, Opioid chemistry, Animals, Dose-Response Relationship, Drug, Mice, Morphinans chemistry, Receptors, Opioid, delta antagonists & inhibitors, Receptors, Opioid, kappa antagonists & inhibitors, Receptors, Opioid, mu antagonists & inhibitors, Thalamus drug effects, Analgesics, Opioid pharmacology, Morphinans pharmacology, Narcotic Antagonists pharmacology, Opioid-Related Disorders drug therapy
- Abstract
The opioid crisis is a significant public health issue with more than 115 people dying from opioid overdose per day in the United States. The aim of the present study was to characterize the in vitro and in vivo pharmacological effects of 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(indole-7-carboxamido)morphinan (NAN), a μ opioid receptor (MOR) ligand that may be a potential candidate for opioid use disorder treatment that produces less withdrawal signs than naltrexone. The efficacy of NAN was compared to varying efficacy ligands at the MOR, and determined at the δ opioid receptor (DOR) and κ opioid receptor (KOR). NAN was identified as a low efficacy partial agonist for G-protein activation at the MOR and DOR, but had relatively high efficacy at the KOR. In contrast to high efficacy MOR agonists, NAN did not induce MOR internalization, downregulation, or desensitization, but it antagonized agonist-induced MOR internalization and stimulation of intracellular Ca
2+ release. Opioid withdrawal studies conducted using morphine-pelleted mice demonstrated that NAN precipitated significantly less withdrawal signs than naltrexone at similar doses. Furthermore, NAN failed to produce fentanyl-like discriminative stimulus effects in rats up to doses that produced dose- and time-dependent antagonism of fentanyl. Overall, these results provide converging lines of evidence that NAN functions mainly as a MOR antagonist and support further consideration of NAN as a candidate medication for opioid use disorder treatment.- Published
- 2019
- Full Text
- View/download PDF
27. Structure-Activity Relationship Studies of 6α- and 6β-Indolylacetamidonaltrexamine Derivatives as Bitopic Mu Opioid Receptor Modulators and Elaboration of the "Message-Address Concept" To Comprehend Their Functional Conversion.
- Author
-
Obeng S, Wang H, Jali A, Stevens DL, Akbarali HI, Dewey WL, Selley DE, and Zhang Y
- Subjects
- Allosteric Regulation drug effects, Allosteric Regulation physiology, Analgesics, Opioid chemistry, Animals, CHO Cells, Cricetinae, Cricetulus, Dose-Response Relationship, Drug, Male, Mice, Narcotic Antagonists chemistry, Protein Binding drug effects, Protein Binding physiology, Protein Structure, Secondary, Receptors, Opioid, mu physiology, Structure-Activity Relationship, Analgesics, Opioid pharmacology, Narcotic Antagonists pharmacology, Receptors, Opioid, mu agonists, Receptors, Opioid, mu antagonists & inhibitors
- Abstract
Structure-activity relationship (SAR) studies of numerous opioid ligands have shown that introduction of a methyl or ethyl group on the tertiary amino group at position 17 of the epoxymorphinan skeleton generally results in a mu opioid receptor (MOR) agonist while introduction of a cyclopropylmethyl group typically leads to an antagonist. Furthermore, it has been shown that introduction of heterocyclic ring systems at position 6 can favor antagonism. However, it was reported that 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-[(2'-indolyl)acetamido]morphinan (INTA), which bears a cyclopropylmethyl group at position 17 and an indole ring at position 6, acted as a MOR agonist. We herein report a SAR study on INTA with a series of its complementary derivatives to understand how introduction of an indole moiety with α or β linkage at position 6 of the epoxymorphinan skeleton may influence ligand function. Interestingly, one of INTA derivatives, compound 15 (NAN) was identified as a MOR antagonist both in vitro and in vivo. Molecular modeling studies revealed that INTA and NAN may interact with different domains of the MOR allosteric binding site. In addition, INTA may interact with W293 and N150 residues found in the orthosteric site to stabilize MOR activation conformation while NAN does not. These results suggest that INTA and NAN may be bitopic ligands and the type of allosteric interactions with the MOR influence their functional activity. These insights along with our enriched comprehension of the "message-address" concept will to benefit future ligand design.
- Published
- 2019
- Full Text
- View/download PDF
28. Design, Synthesis, and Biological Evaluation of the Third Generation 17-Cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-[(4'-pyridyl)carboxamido]morphinan (NAP) Derivatives as μ/κ Opioid Receptor Dual Selective Ligands.
- Author
-
Zheng Y, Obeng S, Wang H, Jali AM, Peddibhotla B, Williams DA, Zou C, Stevens DL, Dewey WL, Akbarali HI, Selley DE, and Zhang Y
- Subjects
- Analgesics, Opioid pharmacology, Animals, Binding Sites, Constipation chemically induced, Constipation drug therapy, Ligands, Male, Mice, Molecular Conformation, Molecular Docking Simulation, Morphinans metabolism, Morphinans therapeutic use, Morphine pharmacology, Receptors, Opioid, kappa genetics, Receptors, Opioid, kappa metabolism, Receptors, Opioid, mu genetics, Receptors, Opioid, mu metabolism, Structure-Activity Relationship, Drug Design, Morphinans chemistry, Receptors, Opioid, kappa antagonists & inhibitors, Receptors, Opioid, mu antagonists & inhibitors
- Abstract
μ opioid receptor (MOR) agonists have been widely applied for treating moderate to severe pain. However, numerous adverse effects have been associated with their application, including opioid-induced constipation (OIC), respiratory depression, and addiction. On the basis of previous work in our laboratory, NAP, a 6β- N-4'-pyridyl substituted naltrexamine derivative, was identified as a peripheral MOR antagonist that may be used to treat OIC. To further explore its structure-activity relationship, a new series of NAP derivatives were designed, synthesized, and biologically evaluated. Among these derivatives, NFP and NYP significantly antagonized the antinociception effect of morphine. Whereas NAP acted mainly peripherally, its derivatives NFP and NYP actually can act centrally. Furthermore, NFP produced significantly lesser withdrawal symptoms than naloxone at similar doses. These results suggest that NFP has the potential to be a lead compound to treat opioid abuse and addiction.
- Published
- 2019
- Full Text
- View/download PDF
29. Methylation Products of 6β- N-Heterocyclic Substituted Naltrexamine Derivatives as Potential Peripheral Opioid Receptor Modulators.
- Author
-
Zheng Y, Obeng S, Wang H, Stevens DL, Komla E, Selley DE, Dewey WL, Akbarali HI, and Zhang Y
- Subjects
- Analgesics, Opioid adverse effects, Animals, CHO Cells, Constipation chemically induced, Constipation drug therapy, Cricetulus, Diarrhea drug therapy, Drug Partial Agonism, Gastrointestinal Agents pharmacology, Gastrointestinal Transit drug effects, Imidazoles pharmacology, Methylation, Mice, Molecular Docking Simulation, Morphine pharmacology, Naltrexone pharmacology, Nociception drug effects, Phenylalanine analogs & derivatives, Phenylalanine pharmacology, Quaternary Ammonium Compounds pharmacology, Radioligand Assay, Receptors, Opioid, delta drug effects, Receptors, Opioid, delta metabolism, Receptors, Opioid, kappa metabolism, Receptors, Opioid, mu metabolism, Analgesics, Opioid pharmacology, Naltrexone analogs & derivatives, Narcotic Antagonists pharmacology, Receptors, Opioid, kappa drug effects, Receptors, Opioid, mu drug effects
- Abstract
Two 6β- N-heterocyclic naltrexamine derivatives, NAP and NMP, have been identified as peripherally selective mu opioid receptor (MOR) antagonists. To further enhance the peripheral selectivity of both compounds, the 17-amino group and the nitrogen atom of the pyridine ring in both NAP and NMP were methylated to obtain dMNAP and dMNMP, respectively. Compared with NAP and NMP, the binding affinities of dMNAP and dMNMP shifted to MOR and KOR (kappa opioid receptor) dual selective and they acted as moderate efficacy partial agonists. The results from radioligand binding studies were further confirmed by molecular docking studies. In vivo studies demonstrated that dMNAP and dMNMP did not produce antinociception nor did they antagonize morphine's antinociceptive activity, indicating that these compounds did not act on the central nervous system. Meanwhile, both dMNAP and dMNMP significantly slowed down fecal excretion, which indicated that they were peripherally acting opioid receptor agonists. All together, these results suggested that dMNAP and dMNMP acted as peripheral mu/kappa opioid receptor modulators and may be applicable in the treatment of diarrhea in patients with bowel dysfunction.
- Published
- 2018
- Full Text
- View/download PDF
30. Early adolescent nicotine exposure affects later-life hippocampal mu-opioid receptors activity and morphine reward but not physical dependence in male mice.
- Author
-
Kota D, Alajaji M, Bagdas D, Selley DE, Sim-Selley LJ, and Damaj MI
- Subjects
- Animals, Hippocampus metabolism, Male, Mice, Mice, Inbred ICR, Nicotine pharmacology, Risk Factors, Hippocampus drug effects, Morphine pharmacology, Nicotine administration & dosage, Receptors, Opioid, mu metabolism, Reward
- Abstract
Rationale: There is extensive literature regarding nicotine-opioid functional interactions. The possibility that use of nicotine products during adolescence might increase the risk of substance abuse such as morphine later in adulthood is particularly relevant to the current opioid crisis., Objectives: To investigate the effects of nicotine exposure for seven days during adolescence in mice on morphine reward as well as morphine physical dependence later in adulthood., Methods: Mice were exposed to nicotine in either early or late adolescence then evaluated for morphine reward and withdrawal symptoms in adulthood. A separate group of mice was exposed to nicotine during adolescent and tissue was evaluated for changes in MOR-mediated G-protein activity using [
35 S]GTPγS binding assays., Results: We report that a 7-day exposure to a low dose of nicotine during early adolescence significantly enhanced morphine preference in the CPP test in adult mice. In contrast, the same treatment with nicotine had no effect on expression of somatic withdrawal signs in morphine-dependent adult mice. MOR-mediated G-protein activity in hippocampus, but not thalamus and striatum of adult mice, was significantly altered by adolescent nicotine treatment., Conclusions: Adolescence is a unique developmental stage during which nicotine has long-term effects on future drug-taking behavior. Further studies are needed to identify the neurotransmitters and mechanisms involved in increased vulnerability to drug abuse., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
31. Differential Tolerance to FTY720-Induced Antinociception in Acute Thermal and Nerve Injury Mouse Pain Models: Role of Sphingosine-1-Phosphate Receptor Adaptation.
- Author
-
Sim-Selley LJ, Wilkerson JL, Burston JJ, Hauser KF, McLane V, Welch SP, Lichtman AH, and Selley DE
- Subjects
- Animals, Disease Models, Animal, Fingolimod Hydrochloride therapeutic use, Male, Mice, Mice, Inbred ICR, Neuralgia physiopathology, Receptors, Lysosphingolipid agonists, Nociceptin, Fingolimod Hydrochloride pharmacology, Neuralgia drug therapy, Neuralgia metabolism, Opioid Peptides drug effects, Receptors, Lysosphingolipid metabolism, Temperature
- Abstract
The immunomodulatory prodrug 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720), which acts as an agonist for sphingosine-1-phosphate (S1P) receptors (S1PR) when phosphorylated, is proposed as a novel pain therapeutic. In this study, we assessed FTY720-mediated antinociception in the radiant heat tail-flick test and in the chronic constriction injury (CCI) model of neuropathic pain in mice. FTY720 produced antinociception and antiallodynia, respectively, and these effects were dose-dependent and mimicked by the S1PR1-selective agonist CYM-5442. Repeated administration of FTY720 for 1 week produced tolerance to acute thermal antinociception, but not to antiallodynia in the CCI model. S1PR-stimulated [
35 S]GTP γ S autoradiography revealed apparent desensitization of G protein activation by S1P or the S1PR1 agonist 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2,4-oxadiazole (SEW-2871) throughout the brain. Similar results were seen in spinal cord membranes, whereby the Emax value of S1PR-stimulated [35 S]GTP γ S binding was greatly reduced in repeated FTY720-treated mice. These results suggest that S1PR1 is a primary target of FTY720 in alleviating both acute thermal nociception and chronic neuropathic nociception. Furthermore, the finding that tolerance develops to antinociception in the tail-flick test but not in chronic neuropathic pain suggests a differential mechanism of FTY720 action between these models. The observation that repeated FTY720 administration led to desensitized S1PR1 signaling throughout the central nervous system suggests the possibility that S1PR1 activation drives the acute thermal antinociceptive effects, whereas S1PR1 desensitization mediates the following: 1) tolerance to thermal antinociceptive actions of FTY720 and 2) the persistent antiallodynic effects of FTY720 in neuropathic pain by producing functional antagonism of pronociceptive S1PR1 signaling., (Copyright © 2018 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2018
- Full Text
- View/download PDF
32. Amphetamine maintenance differentially modulates effects of cocaine, methylenedioxypyrovalerone (MDPV), and methamphetamine on intracranial self-stimulation and nucleus accumbens dopamine in rats.
- Author
-
Johnson AR, Banks ML, Selley DE, and Negus SS
- Subjects
- Animals, Dopamine metabolism, Male, Nucleus Accumbens metabolism, Rats, Sprague-Dawley, Self Stimulation, Serotonin metabolism, Synthetic Cathinone, Amphetamine pharmacology, Benzodioxoles pharmacology, Central Nervous System Agents pharmacology, Cocaine pharmacology, Methamphetamine pharmacology, Nucleus Accumbens drug effects, Pyrrolidines pharmacology
- Abstract
Amphetamine maintenance is effective clinically to reduce the consumption of the monoamine uptake inhibitor cocaine but not of the monoamine releaser methamphetamine, and its effectiveness in treating the abuse of other psychostimulants is not known. The mechanisms for differential amphetamine-maintenance effectiveness to treat different types of psychostimulant abuse are also not known. Accordingly, the present study compared the effects of amphetamine maintenance on abuse-related behavioral and neurochemical effects of cocaine, methamphetamine, and the "bath salts" constituent 3,4-methylenedioxypyrovalerone (MDPV) in rats. In behavioral studies, rats were trained to lever press for electrical brain stimulation in an intracranial self-stimulation (ICSS) procedure. In neurochemical studies, nucleus accumbens (NAc) levels of dopamine (DA) and serotonin (5-HT) were monitored by in vivo microdialysis. Cocaine, methamphetamine, and MDPV each produced dose-dependent ICSS facilitation and increases in NAc DA; cocaine and methamphetamine also increased NAc 5-HT. Amphetamine maintenance (0.32 mg/kg/h × 7 days) produced (1) sustained increases in basal ICSS and NAc DA with no change in NAc 5-HT, (2) blockade of cocaine but not methamphetamine effects on ICSS and NAc DA, and (3) no blockade of cocaine- or methamphetamine-induced increases in NAc 5-HT. Amphetamine maintenance blocked the increases in NAc DA produced by the selective DA uptake inhibitor MDPV, but it did not block MDPV-induced ICSS facilitation. These results show different effects of amphetamine maintenance on behavioral and neurochemical effects of different psychostimulants. The selective effectiveness of amphetamine maintenance to treat cocaine abuse may reflect attenuation of cocaine-induced increases in NAc DA while preserving cocaine-induced increases in NAc 5-HT.
- Published
- 2018
- Full Text
- View/download PDF
33. In vitro and in vivo functional profile characterization of 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3-carboxamido)morphinan (NAQ) as a low efficacy mu opioid receptor modulator.
- Author
-
Obeng S, Yuan Y, Jali A, Selley DE, and Zhang Y
- Subjects
- Animals, CHO Cells, Calcium metabolism, Cricetinae, Cricetulus, beta-Arrestin 2 metabolism, Isoquinolines pharmacology, Receptors, Opioid, mu metabolism
- Abstract
Evidence has shown that downstream signaling by mu opioid receptor (MOR) agonists that recruit β-arrestin2 may lead to the development of tolerance. Also, it has been suggested that opioid receptor desensitization and cyclic AMP overshoot contributes to the development of tolerance and occurrence of withdrawal, respectively. Therefore, studies were conducted with 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3-carboxamido)morphinan (NAQ), a MOR selective partial agonist discovered in our laboratory, to characterize its effect on β-arrestin2 recruitment and precipitation of a cyclic AMP overshoot. DAMGO, a MOR full agonist dose-dependently increased β-arrestin2 association with the MOR, whereas NAQ did not. Moreover, NAQ displayed significant, concentration-dependent antagonism of DAMGO-induced β-arrestin2 recruitment. After prolonged morphine treatment of mMOR-CHO cells, there was a significant overshoot of cAMP upon exposure to naloxone, but not NAQ. Moreover, prolonged incubation of mMOR-CHO cells with NAQ did not result in desensitization nor downregulation of the MOR. In functional studies comparing NAQ with nalbuphine in the cAMP inhibition, Ca
2+ flux and [35 S]GTPγS binding assays, NAQ did not show agonism in the Ca2+ flux assay but showed partial agonism in the cAMP and [35 S]GTPγS assays. Also, NAQ significantly antagonized DAMGO-induced intracellular Ca2+ increase. In conclusion, NAQ is a low efficacy MOR modulator that lacks β-arrestin2 recruitment function and does not induce cellular hallmarks of MOR adaptation and fails to precipitate a cellular manifestation of withdrawal in cells pretreated with morphine. These characteristics are desirable if NAQ is pursued for opioid abuse treatment development., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
34. Δ 9 -Tetrahydrocannabinol Experience Influences ΔFosB and Downstream Gene Expression in Prefrontal Cortex.
- Author
-
Lazenka MF, Kang M, De DD, Selley DE, and Sim-Selley LJ
- Abstract
Introduction: Repeated administration of abused drugs, including Δ
9 -tetrahydrocannabinol (THC), induces the stable transcription factor ΔFosB in dopaminergic terminal field regions of the mesolimbic system. These studies investigated the effect of prior repeated THC treatment on THC-induced ΔFosB expression and regulation of downstream targets in the forebrain. Methods: Mice received THC (10 mg/kg) or vehicle twice daily for 13 days, and then half of each group received a single injection of THC or vehicle 45 min before brain collection. ΔFosB messenger RNA (mRNA) and protein were measured by polymerase chain reaction and immunoblotting, respectively. Potential downstream targets of ΔFosB induction were measured by immunoblot. Results: THC injection in mice with a history of repeated THC treatment enhanced ΔFosB expression as compared with vehicle in the prefrontal cortex (PFC), nucleus accumbens (NAc), and amygdala. This change occurred concomitantly with an increase in ΔFosB mRNA in the PFC and NAc. THC injection in mice with a history of repeated THC treatment increased expression of cyclin-dependent kinase 5 (Cdk5) and its regulatory protein p35 only in the PFC. This increase in Cdk5 and p35 expression in PFC was also found in mice that had only received repeated THC administration, suggesting that this effect might be due to induction of ΔFosB. Extracellular signal-regulated kinase (ERK) phosphorylation was increased in PFC after THC injection in repeated THC-treated mice. Phosphorylation of glycogen synthase kinase-3β (GSK3β), a Cdk5 target, was reduced in PFC after repeated THC treatment regardless of THC history, and phosphorylation of dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) at the Cdk5-regulated threonine 75 site was unchanged. Conclusion: These results suggest that a history of repeated THC administration primes THC-mediated induction of ΔFosB in the NAc and PFC, and that expression of both downstream targets of ΔFosB (e.g., Cdk5 and p35) and upstream activators (e.g., pERK) in the PFC is dependent on THC history, which might have functional implications in addiction and neuropsychiatric disease., Competing Interests: No competing financial interests exist.- Published
- 2017
- Full Text
- View/download PDF
35. Cannabinoid Receptor Interacting Protein 1a Competition with β-Arrestin for CB1 Receptor Binding Sites.
- Author
-
Blume LC, Patten T, Eldeeb K, Leone-Kabler S, Ilyasov AA, Keegan BM, O'Neal JE, Bass CE, Hantgan RR, Lowther WT, Selley DE, and Howlett AL
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Tumor, Green Fluorescent Proteins metabolism, Humans, Membrane Proteins, Peptides chemistry, Phosphorylation, Protein Binding, Rats, Carrier Proteins metabolism, Receptor, Cannabinoid, CB1 metabolism, beta-Arrestins metabolism
- Abstract
Cannabinoid receptor interacting protein 1a (CRIP1a) is a CB
1 receptor (CB1 R) distal C-terminal-associated protein that alters CB1 R interactions with G-proteins. We tested the hypothesis that CRIP1a is capable of also altering CB1 R interactions with β-arrestin proteins that interact with the CB1 R at the C-terminus. Coimmunoprecipitation studies indicated that CB1 R associates in complexes with either CRIP1a or β-arrestin, but CRIP1a and β-arrestin fail to coimmunoprecipitate with each other. This suggests a competition for CRIP1a and β-arrestin binding to the CB1 R, which we hypothesized could attenuate the action of β-arrestin to mediate CB1 R internalization. We determined that agonist-mediated reduction of the density of cell surface endogenously expressed CB1 Rs was clathrin and dynamin dependent and could be modeled as agonist-induced aggregation of transiently expressed GFP-CB1 R. CRIP1a overexpression attenuated CP55940-mediated GFP-CB1 R as well as endogenous β-arrestin redistribution to punctae, and conversely, CRIP1a knockdown augmented β-arrestin redistribution to punctae. Peptides mimicking the CB1 R C-terminus could bind to both CRIP1a in cell extracts as well as purified recombinant CRIP1a. Affinity pull-down studies revealed that phosphorylation at threonine-468 of a CB1 R distal C-terminus 14-mer peptide reduced CB1 R-CRIP1a association. Coimmunoprecipitation of CB1 R protein complexes demonstrated that central or distal C-terminal peptides competed for the CB1 R association with CRIP1a, but that a phosphorylated central C-terminal peptide competed for association with β-arrestin 1, and phosphorylated central or distal C-terminal peptides competed for association with β-arrestin 2. Thus, CRIP1a can compete with β-arrestins for interaction with C-terminal CB1 R domains that could affect agonist-driven, β-arrestin-mediated internalization of the CB1 R., (Copyright © 2016 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2017
- Full Text
- View/download PDF
36. Nanoconjugated NAP as a Potent and Periphery Selective Mu Opioid Receptor Modulator To Treat Opioid-Induced Constipation.
- Author
-
Xu GG, Zolotarskaya OY, Williams DA, Yuan Y, Selley DE, Dewey WL, Akbarali HI, Yang H, and Zhang Y
- Abstract
Opioids are the mainstay for cancer and noncancer pain management. However, their use is often associated with multiple adverse effects. Among them, the most common and persistent one is probably opioid-induced constipation (OIC). Periphery selective opioid antagonists may alleviate the symptoms of OIC without compromising the analgesic effects of opioids. Recently our laboratories have identified one novel lead compound, 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-[(4'-pyridyl)acetamido]morphinan (NAP), as a peripherally selective mu opioid receptor ligand carrying subnanomolar affinity to the mu opioid receptor and over 100-folds of selectivity over both the delta and kappa opioid receptors, with reasonable oral availability and half-life, and potential to treat OIC. Nanoparticle-based drug delivery systems are now widely considered due to their technological advantages such as good stability, high carrier capacity, low therapeutic side effects, etc. Herein we report nanoparticle supported NAP as a potential candidate for OIC treatment with improved peripheral selectivity over the original lead compound NAP.
- Published
- 2016
- Full Text
- View/download PDF
37. Exploration of bivalent ligands targeting putative mu opioid receptor and chemokine receptor CCR5 dimerization.
- Author
-
Arnatt CK, Falls BA, Yuan Y, Raborg TJ, Masvekar RR, El-Hage N, Selley DE, Nicola AV, Knapp PE, Hauser KF, and Zhang Y
- Subjects
- Anti-HIV Agents chemical synthesis, Anti-HIV Agents chemistry, Cyclohexanes chemical synthesis, Cyclohexanes chemistry, Dimerization, Dose-Response Relationship, Drug, HIV Infections virology, HIV-1 drug effects, Humans, Ligands, Maraviroc, Microbial Sensitivity Tests, Models, Molecular, Molecular Structure, Naltrexone chemical synthesis, Naltrexone chemistry, Structure-Activity Relationship, Triazoles chemical synthesis, Triazoles chemistry, Anti-HIV Agents pharmacology, Cyclohexanes pharmacology, HIV Infections drug therapy, Naltrexone pharmacology, Receptors, CCR5 metabolism, Receptors, Opioid, mu antagonists & inhibitors, Triazoles pharmacology
- Abstract
Modern antiretroviral therapies have provided HIV-1 infected patients longer lifespans and better quality of life. However, several neurological complications are now being seen in these patients due to HIV-1 associated injury of neurons by infected microglia and astrocytes. In addition, these effects can be further exacerbated with opiate use and abuse. One possible mechanism for such potentiation effects of opiates is the interaction of the mu opioid receptor (MOR) with the chemokine receptor CCR5 (CCR5), a known HIV-1 co-receptor, to form MOR-CCR5 heterodimer. In an attempt to understand this putative interaction and its relevance to neuroAIDS, we designed and synthesized a series of bivalent ligands targeting the putative CCR5-MOR heterodimer. To understand how these bivalent ligands may interact with the heterodimer, biological studies including calcium mobilization inhibition, binding affinity, HIV-1 invasion, and cell fusion assays were applied. In particular, HIV-1 infection assays using human peripheral blood mononuclear cells, macrophages, and astrocytes revealed a notable synergy in activity for one particular bivalent ligand. Further, a molecular model of the putative CCR5-MOR heterodimer was constructed, docked with the bivalent ligand, and molecular dynamics simulations of the complex was performed in a membrane-water system to help understand the biological observation., (Published by Elsevier Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
38. Stratification of Cannabinoid 1 Receptor (CB1R) Agonist Efficacy: Manipulation of CB1R Density through Use of Transgenic Mice Reveals Congruence between In Vivo and In Vitro Assays.
- Author
-
Grim TW, Morales AJ, Gonek MM, Wiley JL, Thomas BF, Endres GW, Sim-Selley LJ, Selley DE, Negus SS, and Lichtman AH
- Subjects
- Animals, Cannabinoids pharmacology, Female, Guanosine 5'-O-(3-Thiotriphosphate) metabolism, Male, Mice, Mice, Transgenic, Receptor, Cannabinoid, CB1 deficiency, Drug Evaluation, Preclinical methods, Receptor, Cannabinoid, CB1 agonists, Receptor, Cannabinoid, CB1 metabolism
- Abstract
Synthetic cannabinoids (SCs) are an emerging class of abused drugs that differ from each other and the phytocannabinoid ∆
9 -tetrahydrocannabinol (THC) in their safety and cannabinoid-1 receptor (CB1 R) pharmacology. As efficacy represents a critical parameter to understanding drug action, the present study investigated this metric by assessing in vivo and in vitro actions of THC, two well-characterized SCs (WIN55,212-2 and CP55,940), and three abused SCs (JWH-073, CP47,497, and A-834,735-D) in CB1 (+/+), (+/-), and (-/-) mice. All drugs produced maximal cannabimimetic in vivo effects (catalepsy, hypothermia, antinociception) in CB1 (+/+) mice, but these actions were essentially eliminated in CB1 (-/-) mice, indicating a CB1 R mechanism of action. CB1 R efficacy was inferred by comparing potencies between CB1 (+/+) and (+/-) mice [+/+ ED50 /+/- ED50 ], the latter of which has a 50% reduction of CB1 Rs (i.e., decreased receptor reserve). Notably, CB1 (+/-) mice displayed profound rightward and downward shifts in the antinociception and hypothermia dose-response curves of low-efficacy compared with high-efficacy cannabinoids. In vitro efficacy, quantified using agonist-stimulated [35 S]GTPγS binding in spinal cord tissue, significantly correlated with the relative efficacies of antinociception (r = 0.87) and hypothermia (r = 0.94) in CB1 (+/-) mice relative to CB1 (+/+) mice. Conversely, drug potencies for cataleptic effects did not differ between these genotypes and did not correlate with the in vitro efficacy measure. These results suggest that evaluation of antinociception and hypothermia in CB1 transgenic mice offers a useful in vivo approach to determine CB1 R selectivity and efficacy of emerging SCs, which shows strong congruence with in vitro efficacy., (Copyright © 2016 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2016
- Full Text
- View/download PDF
39. Cannabinoid receptor interacting protein suppresses agonist-driven CB 1 receptor internalization and regulates receptor replenishment in an agonist-biased manner.
- Author
-
Blume LC, Leone-Kabler S, Luessen DJ, Marrs GS, Lyons E, Bass CE, Chen R, Selley DE, and Howlett AC
- Subjects
- Animals, Benzoxazines pharmacology, Cell Line, Cell Membrane metabolism, Cyclohexanols pharmacology, Endocannabinoids physiology, Gene Dosage, Gene Knockdown Techniques, Mice, Morpholines pharmacology, Naphthalenes pharmacology, Piperidines pharmacology, Protein Transport, Pyrazoles pharmacology, RNA, Small Interfering, Receptor, Cannabinoid, CB1 genetics, Receptors, Cell Surface drug effects, Rimonabant, Signal Transduction genetics, Carrier Proteins genetics, Carrier Proteins metabolism, Receptor, Cannabinoid, CB1 agonists, Receptor, Cannabinoid, CB1 metabolism
- Abstract
Cannabinoid receptor interacting protein 1a (CRIP1a) is a CB
1 receptor (CB1 R) distal C-terminus-associated protein that modulates CB1 R signaling via G proteins, and CB1 R down-regulation but not desensitization (Blume et al. [2015] Cell Signal., 27, 716-726; Smith et al. [2015] Mol. Pharmacol., 87, 747-765). In this study, we determined the involvement of CRIP1a in CB1 R plasma membrane trafficking. To follow the effects of agonists and antagonists on cell surface CB1 Rs, we utilized the genetically homogeneous cloned neuronal cell line N18TG2, which endogenously expresses both CB1 R and CRIP1a, and exhibits a well-characterized endocannabinoid signaling system. We developed stable CRIP1a-over-expressing and CRIP1a-siRNA-silenced knockdown clones to investigate gene dose effects of CRIP1a on CB1 R plasma membrane expression. Results indicate that CP55940 or WIN55212-2 (10 nM, 5 min) reduced cell surface CB1 R by a dynamin- and clathrin-dependent process, and this was attenuated by CRIP1a over-expression. CP55940-mediated cell surface CB1 R loss was followed by a cycloheximide-sensitive recovery of surface receptors (30-120 min), suggesting the requirement for new protein synthesis. In contrast, WIN55212-2-mediated cell surface CB1 Rs recovered only in CRIP1a knockdown cells. Changes in CRIP1a expression levels did not affect a transient rimonabant (10 nM)-mediated increase in cell surface CB1 Rs, which is postulated to be as a result of rimonabant effects on 'non-agonist-driven' internalization. These studies demonstrate a novel role for CRIP1a in agonist-driven CB1 R cell surface regulation postulated to occur by two mechanisms: 1) attenuating internalization that is agonist-mediated, but not that in the absence of exogenous agonists, and 2) biased agonist-dependent trafficking of de novo synthesized receptor to the cell surface., (© 2016 International Society for Neurochemistry.)- Published
- 2016
- Full Text
- View/download PDF
40. 6β-N-Heterocyclic Substituted Naltrexamine Derivative BNAP: A Peripherally Selective Mixed MOR/KOR Ligand.
- Author
-
Williams DA, Zheng Y, David BG, Yuan Y, Zaidi SA, Stevens DL, Scoggins KL, Selley DE, Dewey WL, Akbarali HI, and Zhang Y
- Subjects
- Animals, Binding, Competitive drug effects, CHO Cells, Cricetulus, Disease Models, Animal, Gastrointestinal Motility drug effects, Guanosine 5'-O-(3-Thiotriphosphate) pharmacokinetics, Guinea Pigs, Ileum anatomy & histology, In Vitro Techniques, Ligands, Male, Mice, Muscle Contraction drug effects, Naltrexone chemical synthesis, Naltrexone chemistry, Neurons drug effects, Receptors, Opioid, kappa metabolism, Receptors, Opioid, mu metabolism, Structure-Activity Relationship, Muscle, Skeletal drug effects, Muscle, Smooth drug effects, Naltrexone analogs & derivatives, Naltrexone pharmacology, Receptors, Opioid, kappa antagonists & inhibitors, Receptors, Opioid, mu antagonists & inhibitors
- Abstract
The 6β-N-heterocyclic naltrexamine derivative, NAP, has been demonstrated to be a peripherally selective mu opioid receptor modulator. To further improve peripheral selectivity of this highly potent ligand, its pyridal ring was quaterinized with benzyl bromide to produce BNAP. In radioligand binding assay, the Ki of BNAP for MOR was 0.76 ± 0.09 nM and was >900-fold more selective for MOR than DOR. The Ki for KOR was 3.46 ± 0.05 nM. In [(35)S]GTPγS ligand stimulated assay, BNAP showed low agonist efficacy with 14.6% of the maximum response of DAMGO with an EC50 of 4.84 ± 0.6 nM. However, unlike its parent compound NAP, BNAP displayed partial agonist activity at KOR with % maximum response at 45.9 ± 1.7% of U50,488H. BNAP did not reverse morphine-induced antinociception when administered subcutaneously but did antagonize when administered intracerebroventricularly. BNAP antagonized morphine-induced contractions of the circular muscle in mice colon. BNAP inhibition of field-stimulated contractions in longitudinal muscle strips for the guinea-pig ileum were also blocked by nor-BNI, a kappa opioid receptor antagonist. BNAP induced inhibition of acetic acid induced abdominal stretching in chronic morphine treated mice. These findings suggest that BNAP is a dual MOR antagonist/KOR agonist and may have functional use in irritable bowel patients.
- Published
- 2016
- Full Text
- View/download PDF
41. Central HIV-1 Tat exposure elevates anxiety and fear conditioned responses of male mice concurrent with altered mu-opioid receptor-mediated G-protein activation and β-arrestin 2 activity in the forebrain.
- Author
-
Hahn YK, Paris JJ, Lichtman AH, Hauser KF, Sim-Selley LJ, Selley DE, and Knapp PE
- Subjects
- AIDS Dementia Complex metabolism, Analgesics, Opioid pharmacology, Animals, Anxiety virology, Conditioning, Psychological physiology, Enkephalin, Ala(2)-MePhe(4)-Gly(5)- pharmacology, GTP-Binding Proteins metabolism, HIV-1, Male, Mice, Mice, Transgenic, Morphine pharmacology, Prosencephalon drug effects, Receptors, Opioid, mu agonists, beta-Arrestin 2 metabolism, tat Gene Products, Human Immunodeficiency Virus genetics, Anxiety metabolism, Fear physiology, Prosencephalon metabolism, Receptors, Opioid, mu metabolism, tat Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
Co-exposure to opiates and HIV/HIV proteins results in enhanced CNS morphological and behavioral deficits in HIV(+) individuals and in animal models. Opiates with abuse liability, such as heroin and morphine, bind preferentially to and have pharmacological actions through μ-opioid-receptors (MORs). The mechanisms underlying opiate-HIV interactions are not understood. Exposure to the HIV-1 transactivator of transcription (Tat) protein causes neurodegenerative outcomes that parallel many aspects of the human disease. We have also observed that in vivo exposure to Tat results in apparent changes in morphine efficacy, and thus have hypothesized that HIV proteins might alter MOR activation. To test our hypothesis, MOR-mediated G-protein activation was determined in neuroAIDS-relevant forebrain regions of transgenic mice with inducible CNS expression of HIV-1 Tat. G-protein activation was assessed by MOR agonist-stimulated [(35)S]guanosine-5'-O-(3-thio)triphosphate ([(35)S]GTPγS) autoradiography in brain sections, and in concentration-effect curves of MOR agonist-stimulated [(35)S]GTPγS binding in membranes isolated from specific brain regions. Comparative studies were done using the MOR-selective agonist DAMGO ([D-Ala(2), N-MePhe(4), Gly-ol]-enkephalin) and a more clinically relevant agonist, morphine. Tat exposure reduced MOR-mediated G-protein activation in an agonist, time, and regionally dependent manner. Levels of the GPCR regulatory protein β-arrestin-2, which is involved in MOR desensitization, were found to be elevated in only one affected brain region, the amygdala; amygdalar β-arrestin-2 also showed a significantly increased association with MOR by co-immunoprecipitation, suggesting decreased availability of MOR. Interestingly, this correlated with changes in anxiety and fear-conditioned extinction, behaviors that have substantial amygdalar input. We propose that HIV-1 Tat alters the intrinsic capacity of MOR to signal in response to agonist binding, possibly via a mechanism involving altered expression and/or function of β-arrestin-2., (Copyright © 2016. Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
42. Discriminative Stimulus Properties of the Endocannabinoid Catabolic Enzyme Inhibitor SA-57 in Mice.
- Author
-
Owens RA, Ignatowska-Jankowska B, Mustafa M, Beardsley PM, Wiley JL, Jali A, Selley DE, Niphakis MJ, Cravatt BF, and Lichtman AH
- Subjects
- Amidohydrolases metabolism, Animals, Cyclohexanols pharmacology, Dose-Response Relationship, Drug, Male, Mice, Mice, Inbred C57BL, Monoacylglycerol Lipases metabolism, Acetamides pharmacology, Amidohydrolases antagonists & inhibitors, Carbamates pharmacology, Discrimination, Psychological drug effects, Endocannabinoids metabolism, Enzyme Inhibitors pharmacology, Monoacylglycerol Lipases antagonists & inhibitors
- Abstract
Whereas the inhibition of fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), the respective major hydrolytic enzymes of N-arachidonoyl ethanolamine (AEA) and 2-arachidonoylglycerol (2-AG), elicits no or partial substitution for Δ(9)-tetrahydrocannabinol (THC) in drug-discrimination procedures, combined inhibition of both enzymes fully substitutes for THC, as well as produces a constellation of cannabimimetic effects. The present study tested whether C57BL/6J mice would learn to discriminate the dual FAAH-MAGL inhibitor SA-57 (4-[2-(4-chlorophenyl)ethyl]-1-piperidinecarboxylic acid 2-(methylamino)-2-oxoethyl ester) from vehicle in the drug-discrimination paradigm. In initial experiments, 10 mg/kg SA-57 fully substituted for CP55,940 ((-)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-trans-4-(3-hydroxypropyl)cyclohexanol), a high-efficacy CB1 receptor agonist in C57BL/6J mice and for AEA in FAAH (-/-) mice. Most (i.e., 23 of 24) subjects achieved criteria for discriminating SA-57 (10 mg/kg) from vehicle within 40 sessions, with full generalization occurring 1 to 2 hours postinjection. CP55,940, the dual FAAH-MAGL inhibitor JZL195 (4-nitrophenyl 4-(3-phenoxybenzyl)piperazine-1-carboxylate), and the MAGL inhibitors MJN110 (2,5-dioxopyrrolidin-1-yl 4-(bis(4-chlorophenyl)methyl)piperazine-1-carboxylate) and JZL184 (4-[Bis(1,3-benzodioxol-5-yl)hydroxymethyl]-1-piperidinecarboxylic acid 4-nitrophenyl ester) fully substituted for SA-57. Although the FAAH inhibitors PF-3845 ((N-3-pyridinyl-4-[[3-[[5-(trifluoromethyl)-2-pyridinyl]oxy]phenyl]methyl]-1-piperidinecarboxamide) and URB597 (cyclohexylcarbamic acid 3'-(aminocarbonyl)-[1,1'-biphenyl]-3-yl ester) did not substitute for SA-57, PF-3845 produced a 2-fold leftward shift in the MJN110 substitution dose-response curve. In addition, the CB1 receptor antagonist rimonabant blocked the generalization of SA-57, as well as substitution of CP55,940, JZL195, MJN110, and JZL184. These findings suggest that MAGL inhibition plays a major role in the CB1 receptor-mediated SA-57 training dose, which is further augmented by FAAH inhibition., (Copyright © 2016 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2016
- Full Text
- View/download PDF
43. Early adolescent nicotine exposure affects later-life cocaine reward in mice.
- Author
-
Alajaji M, Lazenka MF, Kota D, Wise LE, Younis RM, Carroll FI, Levine A, Selley DE, Sim-Selley LJ, and Damaj MI
- Subjects
- Aging psychology, Amphetamine pharmacology, Animals, Central Nervous System Stimulants pharmacology, Conditioning, Operant, Eating drug effects, Male, Mice, Mice, Inbred ICR, Morphine pharmacology, Narcotics pharmacology, Nucleus Accumbens drug effects, Nucleus Accumbens metabolism, Proto-Oncogene Proteins c-fos biosynthesis, Proto-Oncogene Proteins c-fos genetics, Cocaine pharmacology, Cocaine-Related Disorders psychology, Nicotine pharmacology, Nicotinic Agonists pharmacology, Reward
- Abstract
Adolescence represents a unique developmental period associated with increased risk-taking behavior and experimentation with drugs of abuse, in particular nicotine. We hypothesized that exposure to nicotine during early adolescence might increase the risk for drug reward in adulthood. To test this hypothesis, male ICR mice were treated with a subchronic regimen of nicotine or saline during adolescence, and their preference for cocaine, morphine and amphetamine was examined using the conditioned place preference (CPP) test in adulthood. Long-term behavioral changes induced by nicotine suggested a possible role of altered gene transcription. Thus, immunoblot for ΔFosB, a member of the Fos family of transcription factors, was conducted in the nucleus accumbens of these mice. Mice treated with nicotine during early but not late adolescence showed an increase in CPP for cocaine, morphine and amphetamine later in adulthood. This effect was not seen in mice pretreated with a subchronic regimen of nicotine as adults, suggesting that exposure to nicotine specifically during early adolescence increases the rewarding effects of other drugs in adulthood. However, adolescent nicotine exposure did not alter highly palatable food conditioning in mice. The enhancement of cocaine CPP by nicotine was strain-dependent and was blocked by pretreatment with nicotinic antagonists. In addition, nicotine exposure during early adolescence induced ΔFosB expression to a greater extent than identical nicotine exposure in adulthood, and enhanced cocaine-induced locomotor sensitization later in adulthood. These results suggest that nicotine exposure during early adolescence increases drug-induced reward in adulthood through mechanisms that may involve the induction of ΔFosB., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
44. Pharmacological characterization of repeated administration of the first generation abused synthetic cannabinoid CP47,497.
- Author
-
Grim TW, Samano KL, Ignatowska-Jankowska B, Tao Q, Sim-Selly LJ, Selley DE, Wise LE, Poklis A, and Lichtman AH
- Subjects
- Animals, Cannabinoid Receptor Antagonists pharmacology, Dronabinol pharmacology, Drug Tolerance, Endocannabinoids pharmacology, Female, GTP-Binding Proteins metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Inbred ICR, Piperidines pharmacology, Pyrazoles pharmacology, Receptor, Cannabinoid, CB1 antagonists & inhibitors, Rimonabant, Cannabinoids pharmacology
- Abstract
A series of in vivo and in vitro assays were conducted to characterize the pharmacological effects of the first generation abused synthetic cannabinoid CP47,497, a racemic bicyclic cannabinoid that is similar in structure to the potent, high-efficacy synthetic cannabinoid CP55,940. CP47,497 was less efficacious than CP55,940 in activating G-proteins and dose-dependently produced common CB1 receptor-dependent pharmacological effects (i.e. catalepsy, hypothermia, antinociception, and hypolocomotion). CP47,497 also substituted for Δ9-tetrahydrocannabinol (THC) in the mouse drug discrimination, indicating that both drugs elicited a similar interceptive stimulus. The pharmacological effects of CP47,497 underwent tolerance following repeated administration and showed cross-tolerance following repeated THC administration, further suggesting a common cannabimimetic mechanism of action. Finally, the CB1 receptor antagonist rimonabant precipitated similar magnitudes of somatic withdrawal responses in mice treated repeatedly with THC or CP47,497. Taken together, these data verify the acute cannabimimetic effects of CP47,497, and indicate tolerance and dependence following repeated administration. The assays used here provide a straightforward approach to characterize the emerging next generation of abused synthetic cannabinoids.
- Published
- 2016
- Full Text
- View/download PDF
45. 17-Cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-(4'-pyridylcarboxamido)morphinan (NAP) Modulating the Mu Opioid Receptor in a Biased Fashion.
- Author
-
Zhang Y, Williams DA, Zaidi SA, Yuan Y, Braithwaite A, Bilsky EJ, Dewey WL, Akbarali HI, Streicher JM, and Selley DE
- Subjects
- Analgesics, Opioid chemistry, Animals, CHO Cells, Cell Line, Cricetulus, Gastrointestinal Motility drug effects, Humans, Male, Mice, Microscopy, Confocal, Morphinans chemistry, Morphinans pharmacology, Receptors, Opioid, mu metabolism, Analgesics, Opioid pharmacology, Models, Molecular, Receptors, Opioid, mu agonists
- Abstract
Mounting evidence has suggested that G protein-coupled receptors can be stabilized in multiple conformations in response to distinct ligands, which exert discrete functions through selective activation of various downstream signaling events. In accordance with this concept, we report biased signaling of one C6-heterocyclic substituted naltrexamine derivative, namely, 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-(4'-pyridylcarboxamido)morphinan (NAP) at the mu opioid receptor (MOR). NAP acted as a low efficacy MOR partial agonist in the G protein-mediated [(35)S]GTPγS binding assay, whereas it did not significantly induce calcium flux or β-arrestin2 recruitment. In contrast, it potently blocked MOR full agonist-induced β-arrestin2 recruitment and translocation. Additionally, NAP dose-dependently antagonized MOR full agonist-induced intracellular calcium flux and β-arrestin2 recruitment. Further results in an isolated organ bath preparation confirmed that NAP reversed the morphine-induced reduction in colon motility. Ligand docking and dynamics simulation studies of NAP at the MOR provided more supporting evidence for biased signaling of NAP at an atomic level. Due to the fact that NAP is MOR selective and preferentially distributed peripherally upon systemic administration while β-arrestin2 is reportedly required for impairment of intestinal motility by morphine, biased antagonism of β-arrestin2 recruitment by NAP further supports its utility as a treatment for opioid-induced constipation.
- Published
- 2016
- Full Text
- View/download PDF
46. Efficacy of Hybrid Tetrahydrobenzo[d]thiazole Based Aryl Piperazines D-264 and D-301 at D₂ and D₃ Receptors.
- Author
-
Zhen J, Antonio T, Jacob JC, Grandy DK, Reith MEA, Dutta AK, and Selley DE
- Subjects
- Animals, Mice, Mice, Knockout, Piperazines chemistry, Receptors, Dopamine D2 genetics, Thiazoles chemistry, Piperazines pharmacology, Receptors, Dopamine D2 drug effects, Receptors, Dopamine D3 drug effects, Thiazoles pharmacology
- Abstract
In elucidating the role of pharmacodynamic efficacy at D3 receptors in therapeutic effectiveness of dopamine receptor agonists, the influence of study system must be understood. Here two compounds with D3 over D2 selectivity developed in our earlier work, D-264 and D-301, are compared in dopamine receptor-mediated G-protein activation in striatal regions of wild-type and D2 receptor knockout mice and in CHO cells expressing D2 or D3 receptors. In caudate-putamen of D2 knockout mice, D-301 was ~3-fold more efficacious than D-264 in activating G-proteins as assessed by [(35)S]GTPγS binding; in nucleus accumbens, D-301 stimulated G-protein activation whereas D-264 did not. In contrast, the two ligands exerted similar efficacy in both regions of wild-type mice, suggesting both ligands activate D2 receptors with similar efficacy. In D2 and D3 receptor-expressing CHO cells, D-264 and D-301 appeared to act in the [(35)S]GTPγS assay as full agonists because they produced maximal stimulation equal to dopamine. Competition for [(3)H]spiperone binding was then performed to determine Ki/EC50 ratios as an index of receptor reserve for each ligand. Action of D-301, but not D-264, showed receptor reserve in D3 but not in D2 receptor-expressing cells, whereas dopamine showed receptor reserve in both cell lines. Gαo1 is highly expressed in brain and is important in D2-like receptor-G protein coupling. Transfection of Gαo1 in D3- but not D2-expressing CHO cells led to receptor reserve for D-264 without altering receptor expression levels. D-301 and dopamine exhibited receptor reserve in D3-expressing cells both with and without transfection of Gαo1. Altogether, these results indicate that D-301 has greater intrinsic efficacy to activate D3 receptors than D-264, whereas the two compounds act on D2 receptors with similar intrinsic efficacy. These findings also suggest caution in interpreting Emax values from functional assays in receptor-transfected cell models without accounting for receptor reserve.
- Published
- 2016
- Full Text
- View/download PDF
47. Role of Dopamine Type 1 Receptors and Dopamine- and cAMP-Regulated Phosphoprotein Mr 32 kDa in Δ9-Tetrahydrocannabinol-Mediated Induction of ΔFosB in the Mouse Forebrain.
- Author
-
Lazenka MF, Tomarchio AJ, Lichtman AH, Greengard P, Flajolet M, Selley DE, and Sim-Selley LJ
- Subjects
- Amygdala drug effects, Amygdala metabolism, Animals, Dopamine metabolism, Dopamine Antagonists pharmacology, Locomotion drug effects, Locomotion physiology, Male, Mice, Mice, Inbred C57BL, Mice, Inbred ICR, Mice, Knockout, Nucleus Accumbens drug effects, Nucleus Accumbens metabolism, Prefrontal Cortex drug effects, Prefrontal Cortex metabolism, Putamen drug effects, Putamen metabolism, Dopamine and cAMP-Regulated Phosphoprotein 32 metabolism, Dronabinol pharmacology, Prosencephalon drug effects, Prosencephalon metabolism, Proto-Oncogene Proteins c-fos metabolism, Receptors, Dopamine D1 metabolism
- Abstract
Δ(9)-Tetrahydrocannabinol (THC), the main psychoactive component of marijuana, produces motor and motivational effects via interactions with the dopaminergic system in the caudate-putamen and nucleus accumbens. However, the molecular events that underlie these interactions after THC treatment are not well understood. Our study shows that pretreatment with dopamine D1 receptor (D1R) antagonists before repeated administration of THC attenuated induction of Δ FBJ murine osteosarcoma viral oncogene homolog B (ΔFosB) in the nucleus accumbens, caudate-putamen, amygdala, and prefrontal cortex. Anatomical studies showed that repeated THC administration induced ΔFosB in D1R-containing striatal neurons. Dopamine signaling in the striatum involves phosphorylation-specific effects of the dopamine- and cAMP-regulated phosphoprotein Mr 32 kDa (DARPP-32), which regulates protein kinase A signaling. Genetic deletion of DARPP-32 attenuated ΔFosB expression measured after acute, but not repeated, THC administration in both the caudate-putamen and nucleus accumbens. THC was then acutely or repeatedly administered to wild-type (WT) and DARPP-32 knockout (KO) mice, and in vivo responses were measured. DARPP-32 KO mice exhibited enhanced acute THC-mediated hypolocomotion and developed greater tolerance to this response relative to the WT mice. Agonist-stimulated guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPγS) binding showed that cannabinoid-stimulated G-protein activity did not differ between DARPP-32 KO and WT mice treated with vehicle or repeated THC. These results indicate that D1Rs play a major role in THC-mediated ΔFosB induction in the forebrain, whereas the role of DARPP-32 in THC-mediated ΔFosB induction and modulation of motor activity appears to be more complex., (Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
48. Metabolic Interplay between Astrocytes and Neurons Regulates Endocannabinoid Action.
- Author
-
Viader A, Blankman JL, Zhong P, Liu X, Schlosburg JE, Joslyn CM, Liu QS, Tomarchio AJ, Lichtman AH, Selley DE, Sim-Selley LJ, and Cravatt BF
- Subjects
- Animals, Arachidonic Acids genetics, Astrocytes cytology, Endocannabinoids genetics, Glycerides genetics, Mice, Mice, Knockout, Monoacylglycerol Lipases genetics, Monoacylglycerol Lipases metabolism, Neurons cytology, Receptor, Cannabinoid, CB1 genetics, Receptor, Cannabinoid, CB1 metabolism, Arachidonic Acids metabolism, Astrocytes metabolism, Cell Communication physiology, Endocannabinoids metabolism, Glycerides metabolism, Neurons metabolism
- Abstract
The endocannabinoid 2-arachidonoylglycerol (2-AG) is a retrograde lipid messenger that modulates synaptic function, neurophysiology, and behavior. 2-AG signaling is terminated by enzymatic hydrolysis-a reaction that is principally performed by monoacylglycerol lipase (MAGL). MAGL is broadly expressed throughout the nervous system, and the contributions of different brain cell types to the regulation of 2-AG activity in vivo remain poorly understood. Here, we genetically dissect the cellular anatomy of MAGL-mediated 2-AG metabolism in the brain and show that neurons and astrocytes coordinately regulate 2-AG content and endocannabinoid-dependent forms of synaptic plasticity and behavior. We also find that astrocytic MAGL is mainly responsible for converting 2-AG to neuroinflammatory prostaglandins via a mechanism that may involve transcellular shuttling of lipid substrates. Astrocytic-neuronal interplay thus provides distributed oversight of 2-AG metabolism and function and, through doing so, protects the nervous system from excessive CB1 receptor activation and promotes endocannabinoid crosstalk with other lipid transmitter systems., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
49. Full Fatty Acid Amide Hydrolase Inhibition Combined with Partial Monoacylglycerol Lipase Inhibition: Augmented and Sustained Antinociceptive Effects with Reduced Cannabimimetic Side Effects in Mice.
- Author
-
Ghosh S, Kinsey SG, Liu QS, Hruba L, McMahon LR, Grim TW, Merritt CR, Wise LE, Abdullah RA, Selley DE, Sim-Selley LJ, Cravatt BF, and Lichtman AH
- Subjects
- Amidohydrolases metabolism, Animals, Benzodioxoles administration & dosage, Brain drug effects, Brain enzymology, Drug Therapy, Combination, Hyperalgesia drug therapy, Hyperalgesia enzymology, Male, Mice, Mice, Inbred C57BL, Monoacylglycerol Lipases metabolism, Piperidines administration & dosage, Pyridines administration & dosage, Receptor, Cannabinoid, CB1 agonists, Receptor, Cannabinoid, CB1 antagonists & inhibitors, Receptor, Cannabinoid, CB1 metabolism, Time Factors, Treatment Outcome, Amidohydrolases antagonists & inhibitors, Analgesics administration & dosage, Cannabinoid Receptor Agonists administration & dosage, Cannabinoid Receptor Antagonists administration & dosage, Monoacylglycerol Lipases antagonists & inhibitors
- Abstract
Inhibition of fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MAGL), the primary hydrolytic enzymes for the respective endocannabinoids N-arachidonoylethanolamine (AEA) and 2-arachidonylglycerol (2-AG), produces antinociception but with minimal cannabimimetic side effects. Although selective inhibitors of either enzyme often show partial efficacy in various nociceptive models, their combined blockade elicits augmented antinociceptive effects, but side effects emerge. Moreover, complete and prolonged MAGL blockade leads to cannabinoid receptor type 1 (CB1) receptor functional tolerance, which represents another challenge in this potential therapeutic strategy. Therefore, the present study tested whether full FAAH inhibition combined with partial MAGL inhibition would produce sustained antinociceptive effects with minimal cannabimimetic side effects. Accordingly, we tested a high dose of the FAAH inhibitor PF-3845 (N-3-pyridinyl-4-[[3-[[5-(trifluoromethyl)-2-pyridinyl]oxy]phenyl]methyl]-1-piperidinecarboxamide; 10 mg/kg) given in combination with a low dose of the MAGL inhibitor JZL184 [4-nitrophenyl 4-(dibenzo[d][1,3]dioxol-5-yl(hydroxy)methyl)piperidine-1-carboxylate] (4 mg/kg) in mouse models of inflammatory and neuropathic pain. This combination of inhibitors elicited profound increases in brain AEA levels (>10-fold) but only 2- to 3-fold increases in brain 2-AG levels. This combination produced significantly greater antinociceptive effects than single enzyme inhibition and did not elicit common cannabimimetic effects (e.g., catalepsy, hypomotility, hypothermia, and substitution for Δ(9)-tetrahydrocannabinol in the drug-discrimination assay), although these side effects emerged with high-dose JZL184 (i.e., 100 mg/kg). Finally, repeated administration of this combination did not lead to tolerance to its antiallodynic actions in the carrageenan assay or CB1 receptor functional tolerance. Thus, full FAAH inhibition combined with partial MAGL inhibition reduces neuropathic and inflammatory pain states with minimal cannabimimetic effects., (Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
50. Design, syntheses, and pharmacological characterization of 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3'-carboxamido)morphinan analogues as opioid receptor ligands.
- Author
-
Yuan Y, Zaidi SA, Stevens DL, Scoggins KL, Mosier PD, Kellogg GE, Dewey WL, Selley DE, and Zhang Y
- Subjects
- Animals, CHO Cells, Cricetulus, Drug Design, Humans, Isoquinolines therapeutic use, Ligands, Mice, Molecular Dynamics Simulation, Morphinans therapeutic use, Morphine Dependence drug therapy, Morphine Dependence metabolism, Narcotic Antagonists therapeutic use, Receptors, Opioid, mu metabolism, Structure-Activity Relationship, Isoquinolines chemistry, Isoquinolines pharmacology, Morphinans chemistry, Morphinans pharmacology, Narcotic Antagonists chemistry, Narcotic Antagonists pharmacology, Receptors, Opioid, mu antagonists & inhibitors
- Abstract
A series of 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6α-(isoquinoline-3'-carboxamido)morphinan (NAQ) analogues were synthesized and pharmacologically characterized to study their structure-activity relationship at the mu opioid receptor (MOR). The competition binding assay showed two-atom spacer and aromatic side chain were optimal for MOR selectivity. Meanwhile, substitutions at the 1'- and/or 4'-position of the isoquinoline ring retained or improved MOR selectivity over the kappa opioid receptor while still possessing above 20-fold MOR selectivity over the delta opioid receptor. In contrast, substitutions at the 6'- and/or 7'-position of the isoquinoline ring reduced MOR selectivity as well as MOR efficacy. Among this series of ligands, compound 11 acted as an antagonist when challenged with morphine in warm-water tail immersion assay and produced less significant withdrawal symptoms compared to naltrexone in morphine-pelleted mice. Compound 11 also antagonized the intracellular Ca(2+) increase induced by DAMGO. Molecular dynamics simulation studies of 11 in three opioid receptors indicated orientation of the 6'-nitro group varied significantly in the different 'address' domains of the receptors and played a crucial role in the observed binding affinities and selectivity. Collectively, the current findings provide valuable insights for future development of NAQ-based MOR selective ligands., (Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.