1
|
Scherrer KH, Eans SO, Medina JM, Senadheera SN, Khaliq T, Murray TF, McLaughlin JP, Aldrich JV. Tryptophan Substitution in CJ-15,208 ( cyclo[Phe-D-Pro-Phe-Trp]) Introduces δ-Opioid Receptor Antagonism, Preventing Antinociceptive Tolerance and Stress-Induced Reinstatement of Extinguished Cocaine-Conditioned Place Preference. Pharmaceuticals (Basel) 2023; 16:1218. [PMID: 37765026 PMCID: PMC10535824 DOI: 10.3390/ph16091218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The macrocyclic tetrapeptide CJ-15,208 (cyclo[Phe-D-Pro-Phe-Trp]) and its D-Trp isomer exhibit kappa opioid receptor (KOR) antagonism which prevents stress-induced reinstatement of extinguished cocaine-conditioned place preference. Here, we evaluated the effects of substitution of Trp and D-Trp on the peptides' opioid activity, antinociceptive tolerance, and the ability to prevent relapse to extinguished drug-CPP. Six analogs were synthesized using a combination of solid-phase peptide synthesis and cyclization in solution. The analogs were evaluated in vitro for opioid receptor affinity in radioligand competition binding assays, efficacy in the [35S]GTPγS assay, metabolic stability in mouse liver microsomes, and for opioid activity and selectivity in vivo in the mouse 55 °C warm-water tail-withdrawal assay. Potential liabilities of locomotor impairment, respiratory depression, acute tolerance, and conditioned place preference (CPP) were also assessed in vivo, and the ameliorating effect of analogs on the reinstatement of extinguished cocaine-place preference was assessed. Substitutions of other D-amino acids for D-Trp did not affect (or in one case increased) KOR affinity, while two of the three substitutions of an L-amino acid for Trp decreased KOR affinity. In contrast, all but one substitution increased mu opioid receptor (MOR) affinity in vitro. The metabolic stabilities of the analogs were similar to those of their respective parent peptides, with analogs containing a D-amino acid being much more rapidly metabolized than those containing an L-amino acid in this position. In vivo, CJ-15,208 analogs demonstrated antinociception, although potencies varied over an 80-fold range and the mediating opioid receptors differed by substitution. KOR antagonism was lost for all but the D-benzothienylalanine analog, and the 2'-naphthylalanine analog instead demonstrated significant delta opioid receptor (DOR) antagonism. Introduction of DOR antagonism coincided with reduced acute opioid antinociceptive tolerance and prevented stress-induced reinstatement of extinguished cocaine-CPP.
Collapse
Affiliation(s)
- Kristen H. Scherrer
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (K.H.S.); (S.O.E.); (J.M.M.)
| | - Shainnel O. Eans
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (K.H.S.); (S.O.E.); (J.M.M.)
| | - Jessica M. Medina
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (K.H.S.); (S.O.E.); (J.M.M.)
| | - Sanjeewa N. Senadheera
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, USA; (S.N.S.); (T.K.)
| | - Tanvir Khaliq
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, USA; (S.N.S.); (T.K.)
- Department of Medicinal Chemistry, The University of Florida, Gainesville, FL 32610, USA
| | - Thomas F. Murray
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA;
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (K.H.S.); (S.O.E.); (J.M.M.)
| | - Jane V. Aldrich
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045, USA; (S.N.S.); (T.K.)
- Department of Medicinal Chemistry, The University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
2
|
Santino F, Gentilucci L. Design of κ-Opioid Receptor Agonists for the Development of Potential Treatments of Pain with Reduced Side Effects. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010346. [PMID: 36615540 PMCID: PMC9822356 DOI: 10.3390/molecules28010346] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/13/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
The κ-opioid receptor (KOR) has recently emerged as an alternative therapeutic target for the development of pain medications, without deleterious side effects associated with the μ-opioid receptor (MOR). However, modulation of KOR is currently under investigation for the treatment of depression, mood disorders, psychiatric comorbidity, and specific drug addictions. However, KOR agonists also trigger adverse effects including sedation, dysphoria, and hallucinations. In this respect, there is currently much debate on alternative paradigms. Recent effort has been devoted in search of biased ligands capable of selectively activating favorable signaling over signaling associated with unwanted side effects. On the other hand, the use of partial agonists is expected to allow the analgesia to be produced at dosages lower than those required to produce the adverse effects. More empirically, the unwanted central effects can be also avoided by using peripherally restricted agonists. In this review, we discuss the more recent trends in the design of KOR-selective, biased or partial, and finally, peripherally acting agonists. Special emphasis is given on the discussion of the most recent approaches for controlling functional selectivity of KOR-specific ligands.
Collapse
|
3
|
Zhu J, Quizon PM, Wang Y, Adeniran CA, Strauss MJ, Jiménez-Torres AC, Patel P, Cirino TJ, Eans SO, Hammond HR, Deliscar LS, O'Hara P, Saini SK, Ofori E, Vekariya RH, Zhang S, Moukha-Chafiq O, Nguyen TH, Ananthan S, Augelli-Szafran CE, Zhan CG, McLaughlin JP. SRI-32743, a novel allosteric modulator, attenuates HIV-1 Tat protein-induced inhibition of the dopamine transporter and alleviates the potentiation of cocaine reward in HIV-1 Tat transgenic mice. Neuropharmacology 2022; 220:109239. [PMID: 36126727 DOI: 10.1016/j.neuropharm.2022.109239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 08/09/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022]
Abstract
Cocaine abuse increases the incidence of HIV-1-associated neurocognitive disorders. We have demonstrated that HIV-1 transactivator of transcription (Tat) allosterically modulates dopamine (DA) reuptake through the human DA transporter (hDAT), potentially contributing to Tat-induced cognitive impairment and potentiation of cocaine conditioned place preference (CPP). This study determined the effects of a novel allosteric modulator of DAT, SRI-32743, on the interactions of HIV-1 Tat, DA, cocaine, and [3H]WIN35,428 with hDAT in vitro. SRI-32743 (50 nM) attenuated Tat-induced inhibition of [3H]DA uptake and decreased the cocaine-mediated dissociation of [3H]WIN35,428 binding in CHO cells expressing hDAT, suggesting a SRI-32743-mediated allosteric modulation of the Tat-DAT interaction. In further in vivo studies utilizing doxycycline-inducible Tat transgenic (iTat-tg) mice, 14 days of Tat expression significantly reduced the recognition index by 31.7% in the final phase of novel object recognition (NOR) and potentiated cocaine-CPP 2.7-fold compared to responses of vehicle-treated control iTat-tg mice. The Tat-induced NOR deficits and potentiation of cocaine-CPP were not observed in saline-treated iTat-tg or doxycycline-treated G-tg (Tat-null) mice. Systemic administration (i.p.) of SRI-32743 prior to behavioral testing ameliorated Tat-induced impairment of NOR (at a dose of 10 mg/kg) and the Tat-induced potentiation of cocaine-CPP (at doses of 1 or 10 mg/kg). These findings demonstrate that Tat and cocaine interactions with DAT may be regulated by compounds interacting at the DAT allosteric modulatory sites, suggesting a potential therapeutic intervention for HIV-infected patients with concurrent cocaine abuse.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA.
| | - Pamela M Quizon
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Yingying Wang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Charles A Adeniran
- Molecular Modeling and Biopharmaceutical Center, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Matthew J Strauss
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Ana C Jiménez-Torres
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Palak Patel
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Thomas J Cirino
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Haylee R Hammond
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Laure S Deliscar
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Priscilla O'Hara
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| | - Surendra K Saini
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Edward Ofori
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Rakesh H Vekariya
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Sixue Zhang
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Omar Moukha-Chafiq
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Theresa H Nguyen
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | - Subramaniam Ananthan
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL 35205, USA
| | | | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
4
|
Khan MIH, Sawyer BJ, Akins NS, Le HV. A systematic review on the kappa opioid receptor and its ligands: New directions for the treatment of pain, anxiety, depression, and drug abuse. Eur J Med Chem 2022; 243:114785. [PMID: 36179400 DOI: 10.1016/j.ejmech.2022.114785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022]
Abstract
Kappa opioid receptor (KOR) is a member of the opioid receptor system, the G protein-coupled receptors that are expressed throughout the peripheral and central nervous systems and play crucial roles in the modulation of antinociception and a variety of behavioral states like anxiety, depression, and drug abuse. KOR agonists are known to produce potent analgesic effects and have been used clinically for the treatment of pain, while KOR antagonists have shown efficacy in the treatment of anxiety and depression. This review summarizes the history, design strategy, discovery, and development of KOR ligands. KOR agonists are classified as non-biased, G protein-biased, and β-arrestin recruitment-biased, according to their degrees of bias. The mechanisms and associated effects of the G protein signaling pathway and β-arrestin recruitment signaling pathway are also discussed. Meanwhile, KOR antagonists are classified as long-acting and short-acting, based on their half-lives. In addition, we have special sections for mixed KOR agonists and selective peripheral KOR agonists. The mechanisms of action and pharmacokinetic, pharmacodynamic, and behavioral studies for each of these categories are also discussed in this review.
Collapse
Affiliation(s)
- Md Imdadul H Khan
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Benjamin J Sawyer
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Nicholas S Akins
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Hoang V Le
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
5
|
McLaughlin JP, Rayala R, Bunnell AJ, Tantak MP, Eans SO, Nefzi K, Ganno ML, Dooley CT, Nefzi A. Bis-Cyclic Guanidine Heterocyclic Peptidomimetics as Opioid Ligands with Mixed μ-, κ- and δ-Opioid Receptor Interactions: A Potential Approach to Novel Analgesics. Int J Mol Sci 2022; 23:9623. [PMID: 36077029 PMCID: PMC9455983 DOI: 10.3390/ijms23179623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/25/2022] Open
Abstract
The design and development of analgesics with mixed-opioid receptor interactions has been reported to decrease side effects, minimizing respiratory depression and reinforcing properties to generate safer analgesic therapeutics. We synthesized bis-cyclic guanidine heterocyclic peptidomimetics from reduced tripeptides. In vitro screening with radioligand competition binding assays demonstrated variable affinity for the mu-opioid receptor (MOR), delta-opioid receptor (DOR), and kappa-opioid receptor (KOR) across the series, with compound 1968-22 displaying good affinity for all three receptors. Central intracerebroventricular (i.c.v.) administration of 1968-22 produced dose-dependent, opioid receptor-mediated antinociception in the mouse 55 °C warm-water tail-withdrawal assay, and 1968-22 also produced significant antinociception up to 80 min after oral administration (10 mg/kg, p.o.). Compound 1968-22 was detected in the brain 5 min after intravenous administration and was shown to be stable in the blood for at least 30 min. Central administration of 1968-22 did not produce significant respiratory depression, locomotor effects or conditioned place preference or aversion. The data suggest these bis-cyclic guanidine heterocyclic peptidomimetics with multifunctional opioid receptor activity may hold potential as new analgesics with fewer liabilities of use.
Collapse
Affiliation(s)
- Jay P. McLaughlin
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Ramanjaneyulu Rayala
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Ashley J. Bunnell
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Mukund P. Tantak
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Shainnel O. Eans
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Khadija Nefzi
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Michelle L. Ganno
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Colette T. Dooley
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Adel Nefzi
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| |
Collapse
|
6
|
Brice-Tutt AC, Eans SO, Yakovlev D, Aldrich JV, McLaughlin JP. An analog of [d-Trp]CJ-15,208 exhibits kappa opioid receptor antagonism following oral administration and prevents stress-induced reinstatement of extinguished morphine conditioned place preference. Pharmacol Biochem Behav 2022; 217:173405. [PMID: 35584724 DOI: 10.1016/j.pbb.2022.173405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/26/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
Opioid use disorder (OUD) relapse rates are discouragingly high, underscoring the need for new treatment options. The macrocyclic tetrapeptide natural product CJ-15,208 and its stereoisomer [d-Trp]CJ-15,208 demonstrate kappa opioid receptor (KOR) antagonist activity upon oral administration which prevents stress-induced reinstatement of cocaine-seeking behavior. In order to further explore the structure-activity relationships and expand the potential therapeutic applications of KOR antagonism for the treatment of OUD, we screened a series of 24 analogs of [d-Trp]CJ-15,208 with the goal of enhancing KOR antagonist activity. From this screening, analog 22 arose as a compound of interest, demonstrating dose-dependent KOR antagonism after central and oral administration lasting at least 2.5 h. In further oral testing, analog 22 lacked respiratory, locomotor, or reinforcing effects, consistent with the absence of opioid agonism. Pretreatment with analog 22 (30 mg/kg, p.o.) prevented stress-induced reinstatement of extinguished morphine conditioned place preference and reduced some signs of naloxone-precipitated withdrawal in mice physically dependent on morphine. Collectively, these data support the therapeutic potential of KOR antagonists to support abstinence in OUD and ameliorate opioid withdrawal.
Collapse
Affiliation(s)
- Ariana C Brice-Tutt
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Dmitry Yakovlev
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Jane V Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America.
| |
Collapse
|
7
|
Smith MT, Kong D, Kuo A, Imam MZ, Williams CM. Analgesic Opioid Ligand Discovery Based on Nonmorphinan Scaffolds Derived from Natural Sources. J Med Chem 2022; 65:1612-1661. [PMID: 34995453 DOI: 10.1021/acs.jmedchem.0c01915] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Strong opioid analgesics, including morphine, are the mainstays for treating moderate to severe acute pain and alleviating chronic cancer pain. However, opioid-related adverse effects, including nausea or vomiting, sedation, respiratory depression, constipation, pruritus (itch), analgesic tolerance, and addiction and abuse liability, are problematic. In addition, the use of opioids to relieve chronic noncancer pain is controversial due to the "opioid crisis" characterized by opioid misuse or abuse and escalating unintentional death rates due to respiratory depression. Hence, considerable research internationally has been aimed at the "Holy Grail" of the opioid analgesic field, namely the discovery of novel and safer opioid analgesics with improved opioid-related adverse effects. In this Perspective, medicinal chemistry strategies are addressed, where structurally diverse nonmorphinan-based opioid ligands derived from natural sources were deployed as lead molecules. The current state of play, clinical or experimental status, and novel opioid ligand discovery approaches are elaborated in the context of retaining analgesia with improved safety and reduced adverse effects, especially addiction liability.
Collapse
|
8
|
Wilson LL, Eans SO, Ramadan-Siraj I, Modica MN, Romeo G, Intagliata S, McLaughlin JP. Examination of the Novel Sigma-1 Receptor Antagonist, SI 1/28, for Antinociceptive and Anti-allodynic Efficacy against Multiple Types of Nociception with Fewer Liabilities of Use. Int J Mol Sci 2022; 23:615. [PMID: 35054797 PMCID: PMC8775934 DOI: 10.3390/ijms23020615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
Neuropathic pain is a significant problem with few effective treatments lacking adverse effects. The sigma-1 receptor (S1R) is a potential therapeutic target for neuropathic pain, as antagonists for this receptor effectively ameliorate pain in both preclinical and clinical studies. The current research examines the antinociceptive and anti-allodynic efficacy of SI 1/28, a recently reported benzylpiperazine derivative and analog of the S1R antagonist SI 1/13, that was 423-fold more selective for S1R over the sigma-2 receptor (S2R). In addition, possible liabilities of respiration, sedation, and drug reinforcement caused by SI 1/28 have been evaluated. Inflammatory and chemical nociception, chronic nerve constriction injury (CCI) induced mechanical allodynia, and adverse effects of sedation in a rotarod assay, conditioned place preference (CPP), and changes in breath rate and locomotor activity were assessed after i.p. administration of SI 1/28. Pretreatment with SI 1/28 produced dose-dependent antinociception in the formalin test, with an ED50 (and 95% C.I.) value of 13.2 (7.42-28.3) mg/kg, i.p. Likewise, SI 1/28 produced dose-dependent antinociception against visceral nociception and anti-allodynia against CCI-induced neuropathic pain. SI 1/28 demonstrated no impairment of locomotor activity, conditioned place preference, or respiratory depression. In summary, SI 1/28 proved efficacious in the treatment of acute inflammatory pain and chronic neuropathy without liabilities at therapeutic doses, supporting the development of S1R antagonists as therapeutics for chronic pain.
Collapse
Affiliation(s)
- Lisa L. Wilson
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Shainnel O. Eans
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Insitar Ramadan-Siraj
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Maria N. Modica
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| |
Collapse
|
9
|
Aldrich JV, McLaughlin JP. Peptide Kappa Opioid Receptor Ligands and Their Potential for Drug Development. Handb Exp Pharmacol 2022; 271:197-220. [PMID: 34463847 DOI: 10.1007/164_2021_519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ligands for kappa opioid receptors (KOR) have potential uses as non-addictive analgesics and for the treatment of pruritus, mood disorders, and substance abuse. These areas continue to have major unmet medical needs. Significant advances have been made in recent years in the preclinical development of novel opioid peptides, notably ones with structural features that inherently impart stability to proteases. Following a brief discussion of the potential therapeutic applications of KOR agonists and antagonists, this review focuses on two series of novel opioid peptides, all-D-amino acid tetrapeptides as peripherally selective KOR agonists for the treatment of pain and pruritus without centrally mediated side effects, and macrocyclic tetrapeptides based on CJ-15,208 that can exhibit different opioid profiles with potential applications such as analgesics and treatments for substance abuse.
Collapse
Affiliation(s)
- Jane V Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
10
|
The Role of Dynorphin and the Kappa Opioid Receptor in Schizophrenia and Major Depressive Disorder: A Translational Approach. Handb Exp Pharmacol 2021; 271:525-546. [PMID: 33459877 DOI: 10.1007/164_2020_396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kappa opioid receptor (KOR) and its endogenous ligands dynorphins (DYN) have been implicated in the development or symptomatology of a variety of neuropsychiatric disorders. This review covers a brief history of the development of KOR agonists and antagonists, their effects in healthy volunteers, and the potential role of DYN/KOR dysfunction in schizophrenia and major depressive disorder from a translational perspective. The potential role of DYN/KOR dysfunction in schizophrenia is based on several lines of evidence. Selective KOR agonists induce affective states in healthy volunteers with similarities to the symptoms of schizophrenia. Studies have shown increased DYN in patients with schizophrenia, although the data have been mixed. Finally, meta-analytic data have shown that opioid antagonists are associated with reductions in the symptoms of schizophrenia. The potential role of DYN/KOR dysfunction in major depressive disorder is also based on a combination of preclinical and clinical data. Selective KOR agonists have shown pro-depressive effects in human volunteers, while selective KOR antagonists have shown robust efficacy in several preclinical models of antidepressant activity. Small studies have shown that nonselective KOR antagonists may have efficacy in treatment-resistant depression. Additionally, recent clinical data have shown that the KOR may be an effective target for treating anhedonia, a finding relevant to both schizophrenia and depression. Finally, recommendations are provided for translating preclinical models for schizophrenia and major depressive disorder into the clinic.
Collapse
|
11
|
Brice-Tutt AC, Senadheera SN, Ganno ML, Eans SO, Khaliq T, Murray TF, McLaughlin JP, Aldrich JV. Phenylalanine Stereoisomers of CJ-15,208 and [d-Trp]CJ-15,208 Exhibit Distinctly Different Opioid Activity Profiles. Molecules 2020; 25:molecules25173999. [PMID: 32887303 PMCID: PMC7504817 DOI: 10.3390/molecules25173999] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/30/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
The macrocyclic tetrapeptide cyclo[Phe-d-Pro-Phe-Trp] (CJ-15,208) and its stereoisomer cyclo[Phe-d-Pro-Phe-d-Trp] exhibit different opioid activity profiles in vivo. The present study evaluated the influence of the Phe residues’ stereochemistry on the peptides’ opioid activity. Five stereoisomers were synthesized by a combination of solid-phase peptide synthesis and cyclization in solution. The analogs were evaluated in vitro for opioid receptor affinity in radioligand competition binding assays, and for opioid activity and selectivity in vivo in the mouse 55 °C warm-water tail-withdrawal assay. Potential liabilities of locomotor impairment, respiratory depression, acute tolerance development, and place conditioning were also assessed in vivo. All of the stereoisomers exhibited antinociception following either intracerebroventricular or oral administration differentially mediated by multiple opioid receptors, with kappa opioid receptor (KOR) activity contributing for all of the peptides. However, unlike the parent peptides, KOR antagonism was exhibited by only one stereoisomer, while another isomer produced DOR antagonism. The stereoisomers of CJ-15,208 lacked significant respiratory effects, while the [d-Trp]CJ-15,208 stereoisomers did not elicit antinociceptive tolerance. Two isomers, cyclo[d-Phe-d-Pro-d-Phe-Trp] (3) and cyclo[Phe-d-Pro-d-Phe-d-Trp] (5), did not elicit either preference or aversion in a conditioned place preference assay. Collectively, these stereoisomers represent new lead compounds for further investigation in the development of safer opioid analgesics.
Collapse
Affiliation(s)
- Ariana C. Brice-Tutt
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (A.C.B.-T.); (S.O.E.)
| | | | - Michelle L. Ganno
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, FL 34987, USA;
| | - Shainnel O. Eans
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (A.C.B.-T.); (S.O.E.)
| | - Tanvir Khaliq
- Department of Medicinal Chemistry, The University of Florida, Gainesville, FL 32610, USA;
| | - Thomas F. Murray
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA;
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (A.C.B.-T.); (S.O.E.)
- Correspondence: (J.P.M.); (J.V.A.); Tel.: +1-352-273-7207 (J.P.M.); +1-352-273-8708 (J.V.A.)
| | - Jane V. Aldrich
- Department of Medicinal Chemistry, The University of Florida, Gainesville, FL 32610, USA;
- Correspondence: (J.P.M.); (J.V.A.); Tel.: +1-352-273-7207 (J.P.M.); +1-352-273-8708 (J.V.A.)
| |
Collapse
|
12
|
Brice‐Tutt AC, Wilson LL, Eans SO, Stacy HM, Simons CA, Simpson GG, Coleman JS, Ferracane MJ, Aldrich JV, McLaughlin JP. Multifunctional opioid receptor agonism and antagonism by a novel macrocyclic tetrapeptide prevents reinstatement of morphine-seeking behaviour. Br J Pharmacol 2020; 177:4209-4222. [PMID: 32562259 PMCID: PMC7443475 DOI: 10.1111/bph.15165] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 05/16/2020] [Accepted: 06/03/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE The macrocyclic tetrapeptide natural product CJ-15,208 (cyclo[Phe-d-Pro-Phe-Trp]) is a multifunctional μ-opioid receptor and κ-opioid receptor agonist and κ-opioid receptor antagonist that produces antinociception and prevents stress-induced reinstatement of extinguished cocaine-conditioned place preference (CPP). We hypothesized that an analogue of CJ-15,208, cyclo[Pro-Sar-Phe-d-Phe], would demonstrate multifunctional μ-opioid receptor and κ-opioid receptor ligand activity, producing potent antinociception with fewer liabilities than selective μ-opioid receptor agonists, while preventing both drug- and stress-induced reinstatement of morphine-induced CPP. EXPERIMENTAL APPROACH The opioid receptor agonist and antagonist activity of cyclo[Pro-Sar-Phe-d-Phe] was characterized after i.c.v. and i.p. administration to C57BL/6J or transgenic opioid receptor "knockout" mice using the 55°C warm-water tail-withdrawal assay. Liabilities of locomotor coordination, respiration and spontaneous ambulation, and direct rewarding or aversive properties were assessed. Finally, the ability of cyclo[Pro-Sar-Phe-d-Phe] to block morphine- and stress-induced reinstatement of extinguished CPP was determined. KEY RESULTS cyclo[Pro-Sar-Phe-d-Phe] demonstrated dose-dependent, short-lasting antinociception, with an ED50 (and 95% confidence interval) of 0.15 (0.05-0.21) nmol i.c.v. and 1.91 (0.40-3.54) mg·kg-1 i.p., mediated by μ- and κ-opioid receptors. The macrocyclic tetrapeptide also demonstrated potent dose-dependent κ-opioid receptor antagonist-like activity at 2.5, but not at 4.5, h after administration. cyclo[Pro-Sar-Phe-d-Phe] displayed reduced liabiities compared with morphine, attributed to its additional activity at κ-receptors. Pretreatment with cyclo[Pro-Sar-Phe-d-Phe] prevented stress- and drug-induced reinstatement of extinguished morphine-place preference responses in a time-dependent manner. CONCLUSIONS AND IMPLICATIONS These data suggest that cyclo[Pro-Sar-Phe-d-Phe] is a promising lead compound for both the treatment of pain with reduced sideeffects and preventing both drug- and stress-induced relapse in morphine-abstinent subjects.
Collapse
Affiliation(s)
- Ariana C. Brice‐Tutt
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Lisa L. Wilson
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Shainnel O. Eans
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Heather M. Stacy
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Chloe A. Simons
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Grant G. Simpson
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jeremy S. Coleman
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Michael J. Ferracane
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jane V. Aldrich
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
13
|
Norwood V, Brice-Tutt AC, Eans SO, Stacy HM, Shi G, Ratnayake R, Rocca JR, Abboud KA, Li C, Luesch H, McLaughlin JP, Huigens RW. Preventing Morphine-Seeking Behavior through the Re-Engineering of Vincamine's Biological Activity. J Med Chem 2020; 63:5119-5138. [PMID: 31913038 PMCID: PMC7324933 DOI: 10.1021/acs.jmedchem.9b01924] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Indexed: 12/17/2022]
Abstract
Innovative discovery strategies are essential to address the ongoing opioid epidemic in the United States. Misuse of prescription and illegal opioids (e.g., morphine, heroin) has led to major problems with addiction and overdose. We used vincamine, an indole alkaloid, as a synthetic starting point for dramatic structural alterations of its complex, fused ring system to synthesize 80 diverse compounds with intricate molecular architectures. A select series of vincamine-derived compounds were screened for both agonistic and antagonistic activities against a panel of 168 G protein-coupled receptor (GPCR) drug targets. Although vincamine was without an effect, the novel compound 4 (V2a) demonstrated antagonistic activities against hypocretin (orexin) receptor 2. When advanced to animal studies, 4 (V2a) significantly prevented acute morphine-conditioned place preference (CPP) and stress-induced reinstatement of extinguished morphine-CPP in mouse models of opioid reward and relapse. These results demonstrate that the ring distortion of vincamine offers a promising way to explore new chemical space of relevance to opioid addiction.
Collapse
Affiliation(s)
- Verrill
M. Norwood
- Department
of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Ariana C. Brice-Tutt
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Shainnel O. Eans
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Heather M. Stacy
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Guqin Shi
- Department
of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Ranjala Ratnayake
- Department
of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - James R. Rocca
- Department
of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- McKnight
Brain Institute, University of Florida, Gainesville, Florida 32610, United States
| | - Khalil A. Abboud
- Department
of Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Chenglong Li
- Department
of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Hendrik Luesch
- Department
of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Jay P. McLaughlin
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Robert W. Huigens
- Department
of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Center
for Natural Products, Drug Discovery & Development (CNPD3), College
of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
14
|
Ferracane MJ, Brice-Tutt AC, Coleman JS, Simpson GG, Wilson LL, Eans SO, Stacy HM, Murray TF, McLaughlin JP, Aldrich JV. Design, Synthesis, and Characterization of the Macrocyclic Tetrapeptide cyclo[Pro-Sar-Phe-d-Phe]: A Mixed Opioid Receptor Agonist-Antagonist Following Oral Administration. ACS Chem Neurosci 2020; 11:1324-1336. [PMID: 32251585 DOI: 10.1021/acschemneuro.0c00086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Substance abuse remains a serious public health crisis, affecting millions of people worldwide. Macrocyclic tetrapeptides like CJ-15,208 and [d-Trp]CJ-15,208 demonstrate opioid activity shown to attenuate the rewarding effects of cocaine in conditioned place preference assays in mice, making them promising lead compounds for treating substance abuse. In the present study, we report the rational design, synthesis, conformational analysis, and continued pharmacological evaluation of the novel macrocyclic tetrapeptide cyclo[Pro-Sar-Phe-d-Phe] to further explore this unique molecular scaffold. This peptide was rationally designed based on X-ray and NMR structures of related macrocyclic tetrapeptides. Following synthesis, its solution-phase conformations were determined by NMR and molecular modeling. The peptide adopted multiple conformations in polar solvents, but a single conformation in chloroform that is stabilized by intramolecular hydrogen bonding. The peptide is orally bioavailable, producing antinociception and antagonism of kappa opioid receptor (KOR) stimulation following oral administration in a mouse 55 °C warm-water tail-withdrawal assay. Notably, cyclo[Pro-Sar-Phe-d-Phe] blocked both stress- and drug-induced reinstatement of cocaine and morphine conditioned place preference in mice following oral administration, and displayed a decreased side-effect profile compared to morphine. Thus, cyclo[Pro-Sar-Phe-d-Phe] is a promising lead compound for the treatment of substance abuse.
Collapse
Affiliation(s)
- Michael J. Ferracane
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Department of Chemistry, University of Redlands, Redlands, California 92373, United States
| | - Ariana C. Brice-Tutt
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Jeremy S. Coleman
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Grant G. Simpson
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Lisa L. Wilson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Shainnel O. Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Heather M. Stacy
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Thomas F. Murray
- Department of Pharmacology, School of Medicine, Creighton University, Omaha, Nebraska 68178, United States
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Jane V. Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
15
|
Reed B, Butelman ER, Kreek MJ. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Mood and Substance Use Disorders. Handb Exp Pharmacol 2020; 271:473-491. [PMID: 33174064 DOI: 10.1007/164_2020_401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kappa opioid receptor (KOR) and its primary cognate ligands, the dynorphin peptides, are involved in diverse physiological processes. Disruptions to the KOR/dynorphin system have been found to likely play a role in multiple neuropsychological disorders, and hence KOR has emerged as a potential therapeutic target. Targeting KOR is complicated by close homology to the mu and delta opioid receptors (MOR and DOR), and many KOR ligands have at least moderate affinity to MOR and/or DOR. Animal models utilizing primarily very long-lasting selective KOR antagonists (>3 weeks following a single dose) have demonstrated that KOR antagonism attenuates certain anxiety-like and depression-like behaviors and blocks stress- and cue-induced reinstatement to drug seeking. Recently, relatively selective KOR antagonists with medication-like pharmacokinetic and pharmacodynamic properties and durations of action have been developed. One of these, JNJ-67953964 (also referred to as CERC-501, LY2456302, OpraKappa or Aticaprant) has been studied in humans, and shown to be safe, relatively KOR selective, and able to substantially attenuate binding of a KOR PET tracer to CNS localized KOR for greater than 24 h. While animal studies have indicated that compounds of this structural class are capable of normalizing withdrawal signs in animal models of cocaine and alcohol dependence and reducing cocaine and alcohol intake/seeking, additional studies are needed to determine the value of these second generation KOR antagonists in treating mood disorders and substance use disorders in humans.
Collapse
Affiliation(s)
- Brian Reed
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA.
| | - Eduardo R Butelman
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| |
Collapse
|
16
|
Cirino TJ, Eans SO, Medina JM, Wilson LL, Mottinelli M, Intagliata S, McCurdy CR, McLaughlin JP. Characterization of Sigma 1 Receptor Antagonist CM-304 and Its Analog, AZ-66: Novel Therapeutics Against Allodynia and Induced Pain. Front Pharmacol 2019; 10:678. [PMID: 31258480 PMCID: PMC6586922 DOI: 10.3389/fphar.2019.00678] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/24/2019] [Indexed: 12/16/2022] Open
Abstract
Sigma-1 receptors (S1R) and sigma-2 receptors (S2R) may modulate nociception without the liabilities of opioids, offering a promising therapeutic target to treat pain. The purpose of this study was to investigate the in vivo analgesic and anti-allodynic activity of two novel sigma receptor antagonists, the S1R-selective CM-304 and its analog the non-selective S1R/S2R antagonist AZ-66. Inhibition of thermal, induced chemical or inflammatory pain, as well as the allodynia resulting from chronic nerve constriction injury (CCI) and cisplatin exposure as models of neuropathic pain were assessed in male mice. Both sigma receptor antagonists dose-dependently (10–45 mg/kg, i.p.) reduced allodynia in the CCI and cisplatin neuropathic pain models, equivalent at the higher dose to the effect of the control analgesic gabapentin (50 mg/kg, i.p.), although AZ-66 demonstrated a much longer duration of action. Both CM-304 and AZ-66 produced antinociception in the writhing test [0.48 (0.09–1.82) and 2.31 (1.02–4.81) mg/kg, i.p., respectively] equivalent to morphine [1.75 (0.31–7.55) mg/kg, i.p.]. Likewise, pretreatment (i.p.) with either sigma-receptor antagonist dose-dependently produced antinociception in the formalin paw assay of inflammatory pain. However, CM-304 [17.5 (12.7–25.2) mg/kg, i.p.) and AZ-66 [11.6 (8.29–15.6) mg/kg, i.p.) were less efficacious than morphine [3.87 (2.85–5.18) mg/kg, i.p.] in the 55°C warm-water tail-withdrawal assay. While AZ-66 exhibited modest sedative effects in a rotarod assay and conditioned place aversion, CM-304 did not produce significant effects in the place conditioning assay. Overall, these results demonstrate the S1R selective antagonist CM-304 produces antinociception and anti-allodynia with fewer liabilities than established therapeutics, supporting the use of S1R antagonists as potential treatments for chronic pain.
Collapse
Affiliation(s)
- Thomas J Cirino
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Shainnel O Eans
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Jessica M Medina
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Lisa L Wilson
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Marco Mottinelli
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, United States
| | - Sebastiano Intagliata
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, United States
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, United States
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
17
|
Tryptophan-Containing Non-Cationizable Opioid Peptides - a new chemotype with unusual structure and in vivo activity. Future Med Chem 2017; 9:2099-2115. [PMID: 29130348 DOI: 10.4155/fmc-2017-0104] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Recently, a new family of opioid peptides containing tryptophan came to the spotlight for the absence of the fundamental protonable tyramine 'message' pharmacophore. Structure-activity relationship investigations led to diverse compounds, characterized by different selectivity profiles and agonist or antagonist effects. Substitution at the indole of Trp clearly impacted peripheral/central antinociceptivity. These peculiarities prompted to gather all the compounds in a new class, and to coin the definition 'Tryptophan-Containing Non-Cationizable Opioid Peptides', in short 'TryCoNCOPs'. Molecular docking analysis suggested that the TryCoNCOPs can still interact with the receptors in an agonist-like fashion. However, most TryCoNCOPs showed significant differences between the in vitro and in vivo activities, suggesting that opioid activity may be elicited also via alternative mechanisms.
Collapse
|
18
|
Mukhopadhyay A, Hanold LE, Thayele Purayil H, Gisemba SA, Senadheera SN, Aldrich JV. Macrocyclic peptides decrease c-Myc protein levels and reduce prostate cancer cell growth. Cancer Biol Ther 2017; 18:571-583. [PMID: 28692379 PMCID: PMC5652972 DOI: 10.1080/15384047.2017.1345384] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 04/26/2017] [Accepted: 06/17/2017] [Indexed: 10/19/2022] Open
Abstract
The oncoprotein c-Myc is often overexpressed in cancer cells, and the stability of this protein has major significance in deciding the fate of a cell. Thus, targeting c-Myc levels is an attractive approach for developing therapeutic agents for cancer treatment. In this study, we report the anti-cancer activity of the macrocyclic peptides [D-Trp]CJ-15,208 (cyclo[Phe-D-Pro-Phe-D-Trp]) and the natural product CJ-15,208 (cyclo[Phe-D-Pro-Phe-Trp]). [D-Trp]CJ-15,208 reduced c-Myc protein levels in prostate cancer cells and decreased cell proliferation with IC50 values ranging from 2.0 to 16 µM in multiple PC cell lines. [D-Trp]CJ-15,208 induced early and late apoptosis in PC-3 cells following 48 hours treatment, and growth arrest in the G2 cell cycle phase following both 24 and 48 hours treatment. Down regulation of c-Myc in PC-3 cells resulted in loss of sensitivity to [D-Trp]CJ-15,208 treatment, while overexpression of c-Myc in HEK-293 cells imparted sensitivity of these cells to [D-Trp]CJ-15,208 treatment. This macrocyclic tetrapeptide also regulated PP2A by reducing the levels of its phosphorylated form which regulates the stability of cellular c-Myc protein. Thus [D-Trp]CJ-15,208 represents a new lead compound for the potential development of an effective treatment of prostate cancer.
Collapse
Affiliation(s)
- Archana Mukhopadhyay
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas, USA
- Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Laura E. Hanold
- Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Hamsa Thayele Purayil
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, USA
| | - Solomon A. Gisemba
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas, USA
- Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | | | - Jane V. Aldrich
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas, USA
- Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
19
|
Spetea M, Eans SO, Ganno ML, Lantero A, Mairegger M, Toll L, Schmidhammer H, McLaughlin JP. Selective κ receptor partial agonist HS666 produces potent antinociception without inducing aversion after i.c.v. administration in mice. Br J Pharmacol 2017; 174:2444-2456. [PMID: 28494108 PMCID: PMC5513865 DOI: 10.1111/bph.13854] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/09/2017] [Accepted: 05/03/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE The κ receptor has a central role in modulating neurotransmission in central and peripheral neuronal circuits that subserve pain and other behavioural responses. Although κ receptor agonists do not produce euphoria or lead to respiratory suppression, they induce dysphoria and sedation. We hypothesized that brain-penetrant κ receptor ligands possessing biased agonism towards G protein signalling over β-arrestin2 recruitment would produce robust antinociception with fewer associated liabilities. EXPERIMENTAL APPROACH Two new diphenethylamines with high κ receptor selectivity, HS665 and HS666, were assessed following i.c.v. administration in mouse assays of antinociception with the 55°C warm-water tail withdrawal test, locomotor activity in the rotorod and conditioned place preference. The [35 S]-GTPγS binding and β-arrestin2 recruitment in vitro assays were used to characterize biased agonism. KEY RESULTS HS665 (κ receptor agonist) and HS666 (κ receptor partial agonist) demonstrated dose-dependent antinociception after i.c.v. administration mediated by the κ receptor. These highly selective κ receptor ligands displayed varying biased signalling towards G protein coupling in vitro, consistent with a reduced liability profile, reflected by reduced sedation and absence of conditioned place aversion for HS666. CONCLUSIONS AND IMPLICATIONS HS665 and HS666 activate central κ receptors to produce potent antinociception, with HS666 displaying pharmacological characteristics of a κ receptor analgesic with reduced liability for aversive effects correlating with its low efficacy in the β-arrestin2 signalling pathway. Our data provide further understanding of the contribution of central κ receptors in pain suppression, and the prospect of dissociating the antinociceptive effects of HS665 and HS666 from κ receptor-mediated adverse effects.
Collapse
Affiliation(s)
- Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
| | - Shainnel O Eans
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
- Department of PharmacodynamicsUniversity of FloridaGainesvilleFLUSA
| | | | - Aquilino Lantero
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Michael Mairegger
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Lawrence Toll
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Jay P McLaughlin
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
- Department of PharmacodynamicsUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
20
|
De Marco R, Bedini A, Spampinato S, Cavina L, Pirazzoli E, Gentilucci L. Versatile Picklocks To Access All Opioid Receptors: Tuning the Selectivity and Functional Profile of the Cyclotetrapeptide c[Phe-d-Pro-Phe-Trp] (CJ-15,208). J Med Chem 2016; 59:9255-9261. [DOI: 10.1021/acs.jmedchem.6b00420] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Rossella De Marco
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Andrea Bedini
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Irnerio
48, 40126 Bologna, Italy
| | - Santi Spampinato
- Department
of Pharmacy and Biotechnology, University of Bologna, Via Irnerio
48, 40126 Bologna, Italy
| | - Lorenzo Cavina
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Edoardo Pirazzoli
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Luca Gentilucci
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| |
Collapse
|
21
|
Khaliq T, Williams TD, Senadheera SN, Aldrich JV. Development of a robust, sensitive and selective liquid chromatography-tandem mass spectrometry assay for the quantification of the novel macrocyclic peptide kappa opioid receptor antagonist [D-Trp]CJ-15,208 in plasma and application to an initial pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1028:11-15. [PMID: 27318293 DOI: 10.1016/j.jchromb.2016.05.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
Abstract
Selective kappa opioid receptor (KOR) antagonists may have therapeutic potential as treatments for substance abuse and mood disorders. Since [D-Trp]CJ-15,208 (cyclo[Phe-d-Pro-Phe-d-Trp]) is a novel potent KOR antagonist in vivo, it is imperative to evaluate its pharmacokinetic properties to assist the development of analogs as potential therapeutic agents, necessitating the development and validation of a quantitative method for determining its plasma levels. A method for quantifying [D-Trp]CJ-15,208 was developed employing high performance liquid chromatography-tandem mass spectrometry in mouse plasma. Sample preparation was accomplished through a simple one-step protein precipitation method with acetonitrile, and [D-Trp]CJ-15,208 analyzed following HPLC separation on a Hypersil BDS C8 column. Multiple reaction monitoring (MRM), based on the transitions m/z 578.1→217.1 and 245.0, was specific for [D-Trp]CJ-15,208, and MRM based on the transition m/z 566.2→232.9 was specific for the internal standard without interference from endogenous substances in blank mouse plasma. The assay was linear over the concentration range 0.5-500ng/mL with a mean r(2)=0.9987. The mean inter-day accuracy and precision for all calibration standards were 93-118% and 8.9%, respectively. The absolute recoveries were 85±6% and 81±9% for [D-Trp]CJ-15,208 and the internal standard, respectively. The analytical method had excellent sensitivity with a lower limit of quantification of 0.5ng/mL using a sample volume of 20μL. The method was successfully applied to an initial pharmacokinetic study of [D-Trp]CJ-15,208 following intravenous administration to mice.
Collapse
Affiliation(s)
- Tanvir Khaliq
- Department of Medicinal Chemistry, the University of Kansas, Lawrence, KS 66045, USA; Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610, USA
| | - Todd D Williams
- Mass Spectrometry and Analytical Proteomics Laboratory, the University of Kansas, Lawrence, KS 66045, USA
| | - Sanjeewa N Senadheera
- Department of Medicinal Chemistry, the University of Kansas, Lawrence, KS 66045, USA
| | - Jane V Aldrich
- Department of Medicinal Chemistry, the University of Kansas, Lawrence, KS 66045, USA; Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
22
|
Houghten RA, Ganno ML, McLaughlin JP, Dooley C, Eans SO, Santos RG, LaVoi T, Nefzi A, Welmaker G, Giulianotti MA, Toll L. Direct Phenotypic Screening in Mice: Identification of Individual, Novel Antinociceptive Compounds from a Library of 734,821 Pyrrolidine Bis-piperazines. ACS COMBINATORIAL SCIENCE 2016; 18:51-64. [PMID: 26651386 PMCID: PMC4710894 DOI: 10.1021/acscombsci.5b00126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/09/2015] [Indexed: 12/04/2022]
Abstract
The hypothesis in the current study is that the simultaneous direct in vivo testing of thousands to millions of systematically arranged mixture-based libraries will facilitate the identification of enhanced individual compounds. Individual compounds identified from such libraries may have increased specificity and decreased side effects early in the discovery phase. Testing began by screening ten diverse scaffolds as single mixtures (ranging from 17,340 to 4,879,681 compounds) for analgesia directly in the mouse tail withdrawal model. The "all X" mixture representing the library TPI-1954 was found to produce significant antinociception and lacked respiratory depression and hyperlocomotor effects using the Comprehensive Laboratory Animal Monitoring System (CLAMS). The TPI-1954 library is a pyrrolidine bis-piperazine and totals 738,192 compounds. This library has 26 functionalities at the first three positions of diversity made up of 28,392 compounds each (26 × 26 × 42) and 42 functionalities at the fourth made up of 19,915 compounds each (26 × 26 × 26). The 120 resulting mixtures representing each of the variable four positions were screened directly in vivo in the mouse 55 °C warm-water tail-withdrawal assay (ip administration). The 120 samples were then ranked in terms of their antinociceptive activity. The synthesis of 54 individual compounds was then carried out. Nine of the individual compounds produced dose-dependent antinociception equivalent to morphine. In practical terms what this means is that one would not expect multiexponential increases in activity as we move from the all-X mixture, to the positional scanning libraries, to the individual compounds. Actually because of the systematic formatting one would typically anticipate steady increases in activity as the complexity of the mixtures is reduced. This is in fact what we see in the current study. One of the final individual compounds identified, TPI 2213-17, lacked significant respiratory depression, locomotor impairment, or sedation. Our results represent an example of this unique approach for screening large mixture-based libraries directly in vivo to rapidly identify individual compounds.
Collapse
Affiliation(s)
- Richard A. Houghten
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Michelle L. Ganno
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Jay P. McLaughlin
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Colette
T. Dooley
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Shainnel O. Eans
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Radleigh G. Santos
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Travis LaVoi
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Adel Nefzi
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Greg Welmaker
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Marc A. Giulianotti
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Lawrence Toll
- Torrey Pines Institute for
Molecular Studies, 11350
Southwest Village Parkway, Port St. Lucie, Florida 34987, United States
| |
Collapse
|
23
|
Mantsch JR, Baker DA, Funk D, Lê AD, Shaham Y. Stress-Induced Reinstatement of Drug Seeking: 20 Years of Progress. Neuropsychopharmacology 2016; 41:335-56. [PMID: 25976297 PMCID: PMC4677117 DOI: 10.1038/npp.2015.142] [Citation(s) in RCA: 328] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/01/2015] [Accepted: 05/08/2015] [Indexed: 12/24/2022]
Abstract
In human addicts, drug relapse and craving are often provoked by stress. Since 1995, this clinical scenario has been studied using a rat model of stress-induced reinstatement of drug seeking. Here, we first discuss the generality of stress-induced reinstatement to different drugs of abuse, different stressors, and different behavioral procedures. We also discuss neuropharmacological mechanisms, and brain areas and circuits controlling stress-induced reinstatement of drug seeking. We conclude by discussing results from translational human laboratory studies and clinical trials that were inspired by results from rat studies on stress-induced reinstatement. Our main conclusions are (1) The phenomenon of stress-induced reinstatement, first shown with an intermittent footshock stressor in rats trained to self-administer heroin, generalizes to other abused drugs, including cocaine, methamphetamine, nicotine, and alcohol, and is also observed in the conditioned place preference model in rats and mice. This phenomenon, however, is stressor specific and not all stressors induce reinstatement of drug seeking. (2) Neuropharmacological studies indicate the involvement of corticotropin-releasing factor (CRF), noradrenaline, dopamine, glutamate, kappa/dynorphin, and several other peptide and neurotransmitter systems in stress-induced reinstatement. Neuropharmacology and circuitry studies indicate the involvement of CRF and noradrenaline transmission in bed nucleus of stria terminalis and central amygdala, and dopamine, CRF, kappa/dynorphin, and glutamate transmission in other components of the mesocorticolimbic dopamine system (ventral tegmental area, medial prefrontal cortex, orbitofrontal cortex, and nucleus accumbens). (3) Translational human laboratory studies and a recent clinical trial study show the efficacy of alpha-2 adrenoceptor agonists in decreasing stress-induced drug craving and stress-induced initial heroin lapse.
Collapse
Affiliation(s)
- John R Mantsch
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - David A Baker
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Douglas Funk
- Center for Addiction and Mental Health, Campbell Family Mental Health Research Institute, University of Toronto, Toronto, ON, Canada
| | - Anh D Lê
- Center for Addiction and Mental Health, Campbell Family Mental Health Research Institute, University of Toronto, Toronto, ON, Canada
| | - Yavin Shaham
- Intramural Research Program, NIDA-NIH, Baltimore, MD, USA
| |
Collapse
|
24
|
Mediouni S, Jablonski J, Paris JJ, Clementz MA, Thenin-Houssier S, McLaughlin JP, Valente ST. Didehydro-cortistatin A inhibits HIV-1 Tat mediated neuroinflammation and prevents potentiation of cocaine reward in Tat transgenic mice. Curr HIV Res 2015; 13:64-79. [PMID: 25613133 DOI: 10.2174/1570162x13666150121111548] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 11/26/2014] [Accepted: 01/14/2015] [Indexed: 11/22/2022]
Abstract
HIV-1 Tat protein has been shown to have a crucial role in HIV-1-associated neurocognitive disorders (HAND), which includes a group of syndromes ranging from undetectable neurocognitive impairment to dementia. The abuse of psychostimulants, such as cocaine, by HIV infected individuals, may accelerate and intensify neurological damage. On the other hand, exposure to Tat potentiates cocaine-mediated reward mechanisms, which further promotes HAND. Here, we show that didehydro-Cortistatin A (dCA), an analog of a natural steroidal alkaloid, crosses the blood-brain barrier, cross-neutralizes Tat activity from several HIV-1 clades and decreases Tat uptake by glial cell lines. In addition, dCA potently inhibits Tat mediated dysregulation of IL-1β, TNF-α and MCP-1, key neuroinflammatory signaling proteins. Importantly, using a mouse model where doxycycline induces Tat expression, we demonstrate that dCA reverses the potentiation of cocaine-mediated reward. Our results suggest that adding a Tat inhibitor, such as dCA, to current antiretroviral therapy may reduce HIV-1-related neuropathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Susana T Valente
- Department of Infectious diseases, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL 33458, USA.
| |
Collapse
|
25
|
Váradi A, Marrone GF, Eans SO, Ganno ML, Subrath JJ, Le Rouzic V, Hunkele A, Pasternak GW, McLaughlin JP, Majumdar S. Synthesis and characterization of a dual kappa-delta opioid receptor agonist analgesic blocking cocaine reward behavior. ACS Chem Neurosci 2015; 6:1813-24. [PMID: 26325040 DOI: 10.1021/acschemneuro.5b00153] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
3-Iodobenzoyl naltrexamine (IBNtxA) is a potent analgesic belonging to the pharmacologically diverse 6β-amidoepoxymorphinan group of opioids. We present the synthesis and pharmacological evaluation of five analogs of IBNtxA. The scaffold of IBNtxA was modified by removing the 14-hydroxy group, incorporating a 7,8 double bond and various N-17 alkyl substituents. The structural modifications resulted in analogs with picomolar affinities for opioid receptors. The lead compound (MP1104) was found to exhibit approximately 15-fold greater antinociceptive potency (ED50 = 0.33 mg/kg) compared with morphine, mediated through the activation of kappa- and delta-opioid receptors. Despite its kappa agonism, this lead derivative did not cause place aversion or preference in mice in a place-conditioning assay, even at doses 3 times the analgesic ED50. However, pretreatment with the lead compound prevented the reward behavior associated with cocaine in a conditioned place preference assay. Together, these results suggest the promise of dual acting kappa- and delta-opioid receptor agonists as analgesics and treatments for cocaine addiction.
Collapse
Affiliation(s)
- András Váradi
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Gina F. Marrone
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Shainnel O. Eans
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Michelle L. Ganno
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Joan J. Subrath
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Valerie Le Rouzic
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Amanda Hunkele
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Gavril W. Pasternak
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Jay P. McLaughlin
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Susruta Majumdar
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| |
Collapse
|
26
|
Almatroudi A, Husbands SM, Bailey CP, Bailey SJ. Combined administration of buprenorphine and naltrexone produces antidepressant-like effects in mice. J Psychopharmacol 2015; 29:812-21. [PMID: 26045511 PMCID: PMC5075030 DOI: 10.1177/0269881115586937] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Opiates have been used historically for the treatment of depression. Renewed interest in the use of opiates as antidepressants has focused on the development of kappa opioid receptor (κ-receptor) antagonists. Buprenorphine acts as a partial µ-opioid receptor agonist and a κ-receptor antagonist. By combining buprenorphine with the opioid antagonist naltrexone, the activation of µ-opioid receptors will be reduced and the κ-antagonist properties enhanced. We have established that a combination dose of buprenorphine (1 mg/kg) with naltrexone (1 mg/kg) functions as a short-acting κ-antagonist in the mouse tail withdrawal test. Furthermore, this dose combination is neither rewarding nor aversive in the conditioned place preference paradigm, and is without significant locomotor effects. We have shown for the first time that systemic co-administration of buprenorphine (1 mg/kg) with naltrexone (1 mg/kg) in CD-1 mice produced an antidepressant-like response in behaviours in both the forced swim test and novelty induced hypophagia task. Behaviours in the elevated plus maze and light dark box were not significantly altered by treatment with buprenorphine alone, or in combination with naltrexone. We propose that the combination of buprenorphine with naltrexone represents a novel, and potentially a readily translatable approach, to the treatment of depression.
Collapse
Affiliation(s)
| | | | | | - Sarah J Bailey
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| |
Collapse
|
27
|
Eans SO, Ganno ML, Mizrachi E, Houghten RA, Dooley CT, McLaughlin JP, Nefzi A. Parallel Synthesis of Hexahydrodiimidazodiazepines Heterocyclic Peptidomimetics and Their in Vitro and in Vivo Activities at μ (MOR), δ (DOR), and κ (KOR) Opioid Receptors. J Med Chem 2015; 58:4905-17. [PMID: 25996309 DOI: 10.1021/jm501637c] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In the development of analgesics with mixed-opioid agonist activity, peripherally selective activity is expected to decrease side effects, minimizing respiratory depression and reinforcing properties generating significantly safer analgesic therapeutics. We synthesized diazaheterocyclics from reduced tripeptides. In vitro screening with radioligand competition binding assays demonstrated variable affinity for μ (MOR), δ (DOR), and κ (KOR) opioid receptors across the series, with the diimidazodiazepine 14 (2065-14) displaying good affinity for DOR and KOR. Central (icv), intraperitoneal (ip), or oral (po) administration of 14 produced dose-dependent, opioid-receptor mediated antinociception in the mouse, as determined from a 55 °C warm-water tail-withdrawal assay. Only trace amounts of compound 14 was found in brain up to 90 min later, suggesting poor BBB penetration and possible peripherally restricted activity. Central administration of 14 did not produce locomotor effects, acute antinociceptive tolerance, or conditioned-place preference or aversion. The data suggest these diazaheterocyclic mixed activity opioid receptor agonists may hold potential as new analgesics with fewer liabilities of use.
Collapse
Affiliation(s)
- Shainnel O Eans
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Michelle L Ganno
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Elisa Mizrachi
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Richard A Houghten
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Colette T Dooley
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Jay P McLaughlin
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Adel Nefzi
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| |
Collapse
|
28
|
Sedki F, Eigenmann K, Gelinas J, Schouela N, Courchesne S, Shalev U. A role for kappa-, but not mu-opioid, receptor activation in acute food deprivation-induced reinstatement of heroin seeking in rats. Addict Biol 2015; 20:423-32. [PMID: 24725195 DOI: 10.1111/adb.12133] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stress is considered to be one of the major triggers to drug relapse, even after prolonged periods of abstinence. In rats, the activation of stress-related brain systems, including corticotropin-releasing factor and norepinephrine, is critical for stress-induced reinstatement of extinguished drug seeking, an animal model for drug relapse. In addition, there are strong indications that activation of the endogenous opioid system is important for the effects of stress on drug seeking. More specifically, activation of the dynorphin/kappa opioid receptor (KOR) system is critically involved in the reinstatement of cocaine seeking following exposure to stressors, such as footshock, forced swimming or social stress. However, studies on the role of the dynorphin/KOR system in stress-induced reinstatement of heroin seeking are scarce. Here, rats were trained to self-administer heroin (0.1 mg/kg/infusion) for 10 days. Drug seeking was then extinguished and the rats were tested for acute (21 hours) food deprivation-induced reinstatement of heroin seeking. In two separate experiments, rats were injected with the mu-opioid receptor (MOR) antagonist, naltrexone (0.0, 1.0, 10.0 mg/kg; s.c.) or the KOR antagonist, norBNI (0.0, 1.0, 10.0 mg/kg; i.p.) before the reinstatement test. Naltrexone treatment did not affect stress-induced reinstatement. In contrast, treatment with norBNI dose-dependently attenuated food deprivation-induced reinstatement of heroin seeking. These results support the hypothesis that activation of KOR, but not MOR, is critically involved in stress-induced reinstatement of drug seeking.
Collapse
Affiliation(s)
- Firas Sedki
- Department of Psychology; Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie Comportementale; Concordia University; Montreal Quebec Canada
| | - Karine Eigenmann
- Department of Psychology; Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie Comportementale; Concordia University; Montreal Quebec Canada
| | - Jessica Gelinas
- Department of Psychology; Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie Comportementale; Concordia University; Montreal Quebec Canada
| | - Nicholas Schouela
- Department of Psychology; Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie Comportementale; Concordia University; Montreal Quebec Canada
| | - Shannon Courchesne
- Department of Psychology; Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie Comportementale; Concordia University; Montreal Quebec Canada
| | - Uri Shalev
- Department of Psychology; Center for Studies in Behavioral Neurobiology/Groupe de Recherche en Neurobiologie Comportementale; Concordia University; Montreal Quebec Canada
| |
Collapse
|
29
|
Synthesis and biological evaluations of novel endomorphin analogues containing α-hydroxy-β-phenylalanine (AHPBA) displaying mixed μ/δ opioid receptor agonist and δ opioid receptor antagonist activities. Eur J Med Chem 2015; 92:270-81. [DOI: 10.1016/j.ejmech.2014.12.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 12/28/2014] [Indexed: 12/29/2022]
|
30
|
Aldrich JV, Senadheera SN, Ross NC, Reilley KA, Ganno ML, Eans SE, Murray TF, McLaughlin JP. Alanine analogues of [D-Trp]CJ-15,208: novel opioid activity profiles and prevention of drug- and stress-induced reinstatement of cocaine-seeking behaviour. Br J Pharmacol 2015; 171:3212-22. [PMID: 24588614 DOI: 10.1111/bph.12664] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 01/30/2014] [Accepted: 02/24/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE The novel macrocyclic peptide cyclo[Phe-D-Pro-Phe-D-Trp] ([D-Trp]CJ-15,208) exhibits κ opioid (KOP) receptor antagonist activity in both in vitro and in vivo assays. The four alanine analogues of this peptide were synthesized and characterized both in vitro and in vivo to assess the contribution of different amino acid residues to the activity of [D-Trp]CJ-15,208. EXPERIMENTAL APPROACH The peptides were synthesized by a combination of solid phase peptide synthesis and cyclization in solution. The analogues were evaluated in vitro in receptor binding and functional assays, and in vivo with mice using a tail-withdrawal assay for antinociceptive and opioid antagonist activity. Mice demonstrating extinction of cocaine conditioned-place preference (CPP) were pretreated with selected analogues to evaluate prevention of stress or cocaine-induced reinstatement of CPP. KEY RESULTS The alanine analogues displayed pharmacological profiles in vivo distinctly different from [D-Trp]CJ-15,208. While the analogues exhibited varying opioid receptor affinities and κ and μ opioid receptor antagonist activity in vitro, they produced potent opioid receptor-mediated antinociception (ED50 = 0.28-4.19 nmol, i.c.v.) in vivo. Three of the analogues also displayed KOP receptor antagonist activity in vivo. Pretreatment with an analogue exhibiting both KOP receptor agonist and antagonist activity in vivo prevented both cocaine- and stress-induced reinstatement of cocaine-seeking behaviour in the CPP assay in a time-dependent manner. CONCLUSIONS AND IMPLICATIONS These unusual macrocyclic peptides exhibit in vivo opioid activity profiles different from the parent compound and represent novel compounds for potential development as therapeutics for drug abuse and possibly as analgesics.
Collapse
Affiliation(s)
- J V Aldrich
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Rational Approach to the Design of Bioactive Peptidomimetics: Recent Developments in Opioid Agonist Peptides. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2015. [DOI: 10.1016/b978-0-444-63462-7.00002-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
32
|
Urbano M, Guerrero M, Rosen H, Roberts E. Antagonists of the kappa opioid receptor. Bioorg Med Chem Lett 2014; 24:2021-32. [PMID: 24690494 DOI: 10.1016/j.bmcl.2014.03.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/11/2014] [Accepted: 03/15/2014] [Indexed: 01/26/2023]
Abstract
The research community has increasingly focused on the development of OPRK antagonists as pharmacotherapies for the treatment of depression, anxiety, addictive disorders and other psychiatric conditions produced or exacerbated by stress. Short-acting OPRK antagonists have been recently developed as a potential improvement over long-acting prototypic ligands including nor-BNI and JDTic. Remarkably the short-acting LY2456302 is undergoing phase II clinical trials for the augmentation of the antidepressant therapy in treatment-resistant depression. This Letter reviews relevant chemical and pharmacological advances in the identification and development of OPRK antagonists.
Collapse
Affiliation(s)
- Mariangela Urbano
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Miguel Guerrero
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Hugh Rosen
- Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States; The Scripps Research Institute Molecular Screening Center, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States; Department of Immunology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Edward Roberts
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States.
| |
Collapse
|
33
|
Casal-Dominguez JJ, Furkert D, Ostovar M, Teintang L, Clark MJ, Traynor JR, Husbands SM, Bailey SJ. Characterization of BU09059: a novel potent selective κ-receptor antagonist. ACS Chem Neurosci 2014; 5:177-84. [PMID: 24410326 DOI: 10.1021/cn4001507] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Kappa-opioid receptor (κ) antagonists are potential therapeutic agents for a range of psychiatric disorders. The feasibility of developing κ-antagonists has been limited by the pharmacodynamic properties of prototypic κ-selective antagonists; that is, they inhibit receptor signaling for weeks after a single administration. To address this issue, novel trans-(3R,4R)-dimethyl-4-(3-hydroxyphenyl) piperidine derivatives, based on JDTic, were designed using soft-drug principles. The aim was to determine if the phenylpiperidine-based series of κ-antagonists was amenable to incorporation of a potentially metabolically labile group, while retaining good affinity and selectivity for the κ-receptor. Opioid receptor binding affinity and selectivity of three novel compounds (BU09057, BU09058, and BU09059) were tested. BU09059, which most closely resembles JDTic, had nanomolar affinity for the κ-receptor, with 15-fold and 616-fold selectivity over μ- and δ-receptors, respectively. In isolated tissues, BU09059 was a potent and selective κ-antagonist (pA2 8.62) compared with BU09057 (pA2 6.87) and BU09058 (pA2 6.76) which were not κ-selective. In vivo, BU09059 (3 and 10 mg/kg) significantly blocked U50,488-induced antinociception and was as potent as, but shorter acting than, the prototypic selective κ-antagonist norBNI. These data show that a new JDTic analogue, BU09059, retains high affinity and selectivity for the κ-receptor and has a shorter duration of κ-antagonist action in vivo.
Collapse
Affiliation(s)
| | - Daniel Furkert
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Mehrnoosh Ostovar
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Linnea Teintang
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Mary J. Clark
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John R. Traynor
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Stephen. M. Husbands
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| | - Sarah J. Bailey
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, United Kingdom
| |
Collapse
|
34
|
Bidlack JM. Mixed κ/μ partial opioid agonists as potential treatments for cocaine dependence. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:387-418. [PMID: 24484983 DOI: 10.1016/b978-0-12-420118-7.00010-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cocaine use activates the dopamine reward pathway, leading to the reinforcing effects of dopamine. There is no FDA-approved medication for treating cocaine dependence. Opioid agonists and antagonists have been approved for treating opioid and alcohol dependence. Agonists that activate the μ opioid receptor increase dopamine levels in the nucleus accumbens, while μ receptor antagonists decrease dopamine levels by blocking the effects of endogenous opioid peptides. Activation of the κ opioid receptor decreases dopamine levels and leads to dysphoria. In contrast, inhibition of the κ opioid receptor decreases dopamine levels in the nucleus accumbens. Antagonists acting at the κ receptor reduce stress-mediated behaviors and anxiety. Mixed partial μ/κ agonists have the potential of striking a balance between dopamine levels and attenuating relapse to cocaine. The pharmacological properties of mixed μ/κ opioid receptor agonists will be discussed and results from clinical and preclinical studies will be presented. Results from studies with some of the classical benzomorphans and morphinans will be presented as they lay the foundation for structure-activity relationships. Recent results with other partial opioid agonists, including buprenorphine derivatives and the mixed μ/κ peptide CJ-15,208, will be discussed. The behavioral effects of the mixed μ/κ MCL-741, an aminothiazolomorphinan, in attenuating cocaine-induced locomotor activity will be presented. While not a mixed μ/κ opioid, results obtained with GSK1521498, a μ receptor inverse agonist, will be discussed. Preclinical strategies and successes will lay the groundwork for the further development of mixed μ/κ opioid receptor agonists to treat cocaine dependence.
Collapse
Affiliation(s)
- Jean M Bidlack
- School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA.
| |
Collapse
|
35
|
Van't Veer A, Carlezon WA. Role of kappa-opioid receptors in stress and anxiety-related behavior. Psychopharmacology (Berl) 2013; 229:435-52. [PMID: 23836029 PMCID: PMC3770816 DOI: 10.1007/s00213-013-3195-5] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/17/2013] [Indexed: 12/15/2022]
Abstract
RATIONALE Accumulating evidence indicates that brain kappa-opioid receptors (KORs) and dynorphin, the endogenous ligand that binds at these receptors, are involved in regulating states of motivation and emotion. These findings have stimulated interest in the development of KOR-targeted ligands as therapeutic agents. As one example, it has been suggested that KOR antagonists might have a wide range of indications, including the treatment of depressive, anxiety, and addictive disorders, as well as conditions characterized by co-morbidity of these disorders (e.g., post-traumatic stress disorder) A general effect of reducing the impact of stress may explain how KOR antagonists can have efficacy in such a variety of animal models that would appear to represent different disease states. OBJECTIVE Here, we review evidence that disruption of KOR function attenuates prominent effects of stress. We will describe behavioral and molecular endpoints including those from studies that characterize the effects of KOR antagonists and KOR ablation on the effects of stress itself, as well as on the effects of exogenously delivered corticotropin-releasing factor, a brain peptide that mediates key effects of stress. CONCLUSION Collectively, available data suggest that KOR disruption produces anti-stress effects and under some conditions can prevent the development of stress-induced adaptations. As such, KOR antagonists may have unique potential as therapeutic agents for the treatment and even prevention of stress-related psychiatric illness, a therapeutic niche that is currently unfilled.
Collapse
MESH Headings
- Animals
- Anti-Anxiety Agents/pharmacology
- Anti-Anxiety Agents/therapeutic use
- Anxiety Disorders/drug therapy
- Anxiety Disorders/metabolism
- Anxiety Disorders/psychology
- Behavior, Animal/drug effects
- Brain/drug effects
- Brain/metabolism
- Brain/physiopathology
- Corticotropin-Releasing Hormone/metabolism
- Dynorphins/genetics
- Dynorphins/metabolism
- Humans
- Ligands
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
- Stress, Psychological/psychology
Collapse
Affiliation(s)
- Ashlee Van't Veer
- Department of Psychiatry, Harvard Medical School, McLean Hospital, MRC 217, 115 Mill Street, Belmont, MA, 02478, USA
| | | |
Collapse
|