201
|
Vekariya RH, Lei W, Ray A, Saini SK, Zhang S, Molnar G, Barlow D, Karlage KL, Bilsky EJ, Houseknecht KL, Largent-Milnes TM, Streicher JM, Ananthan S. Synthesis and Structure–Activity Relationships of 5′-Aryl-14-alkoxypyridomorphinans: Identification of a μ Opioid Receptor Agonist/δ Opioid Receptor Antagonist Ligand with Systemic Antinociceptive Activity and Diminished Opioid Side Effects. J Med Chem 2020; 63:7663-7694. [DOI: 10.1021/acs.jmedchem.0c00503] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Rakesh H. Vekariya
- Chemistry Department, Southern Research, Birmingham, Alabama 35205, United States
| | - Wei Lei
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Abhisek Ray
- Chemistry Department, Southern Research, Birmingham, Alabama 35205, United States
| | - Surendra K. Saini
- Chemistry Department, Southern Research, Birmingham, Alabama 35205, United States
| | - Sixue Zhang
- Chemistry Department, Southern Research, Birmingham, Alabama 35205, United States
| | - Gabriella Molnar
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Deborah Barlow
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005, United States
| | - Kelly L. Karlage
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Edward J. Bilsky
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Karen L. Houseknecht
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005, United States
| | - Tally M. Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - John M. Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Subramaniam Ananthan
- Chemistry Department, Southern Research, Birmingham, Alabama 35205, United States
| |
Collapse
|
202
|
Grinnell SG, Uprety R, Varadi A, Subrath J, Hunkele A, Pan YX, Pasternak GW, Majumdar S. Synthesis and Characterization of Azido Aryl Analogs of IBNtxA for Radio-Photoaffinity Labeling Opioid Receptors in Cell Lines and in Mouse Brain. Cell Mol Neurobiol 2020; 41:977-993. [PMID: 32424771 DOI: 10.1007/s10571-020-00867-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/05/2020] [Indexed: 10/24/2022]
Abstract
Mu opioid receptors (MOR-1) mediate the biological actions of clinically used opioids such as morphine, oxycodone, and fentanyl. The mu opioid receptor gene, OPRM1, undergoes extensive alternative splicing, generating multiple splice variants. One type of splice variants are truncated variants containing only six transmembrane domains (6TM) that mediate the analgesic action of novel opioid drugs such as 3'-iodobenzoylnaltrexamide (IBNtxA). Previously, we have shown that IBNtxA is a potent analgesic effective in a spectrum of pain models but lacks many side-effects associated with traditional opiates. In order to investigate the targets labeled by IBNtxA, we synthesized two arylazido analogs of IBNtxA that allow photolabeling of mouse mu opioid receptors (mMOR-1) in transfected cell lines and mMOR-1 protein complexes that may comprise the 6TM sites in mouse brain. We demonstrate that both allyl and alkyne arylazido derivatives of IBNtxA efficiently radio-photolabeled mMOR-1 in cell lines and MOR-1 protein complexes expressed either exogenously or endogenously, as well as found in mouse brain. In future, design and application of such radio-photolabeling ligands with a conjugated handle will provide useful tools for further isolating or purifying MOR-1 to investigate site specific ligand-protein contacts and its signaling complexes.
Collapse
Affiliation(s)
- Steven G Grinnell
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA. .,Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| | - Rajendra Uprety
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Andras Varadi
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Joan Subrath
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Amanda Hunkele
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Ying Xian Pan
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Gavril W Pasternak
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.,Pharmacology Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Susruta Majumdar
- Center for Clinical Pharmacology, St Louis College of Pharmacy and Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
203
|
Piekielna-Ciesielska J, Wtorek K, Janecka A. Biased Agonism as an Emerging Strategy in the Search for Better Opioid Analgesics. Curr Med Chem 2020; 27:1562-1575. [PMID: 31057099 DOI: 10.2174/0929867326666190506103124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 12/12/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
Abstract
Morphine and related drugs that act through activating opioid receptors are the most effective analgesics for the relief of severe pain. They have been used for decades, despite the range of unwanted side effects that they produce, as no alternative has been found so far. The major goal of opioid research is to understand the mechanism of action of opioid receptor agonists and to improve the therapeutic utility of opioid drugs. In the search for safer and more potent analgesics, analogs with mixed opioid receptor profile gained a lot of interest. However, recently the concept of biased agonism, that highlights the fact that some ligands are able to differentially activate receptor downstream pathways, became a new approach in the design of novel drug candidates for clinical application. In this review, we summarize current knowledge on the development of opioid ligands of peptide and nonpeptide structure, showing how much opioid pharmacology evolved in recent years.
Collapse
Affiliation(s)
| | - Karol Wtorek
- Department of Biomolecular Chemistry, Medical University, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University, Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
204
|
Eiselt É, Otis V, Belleville K, Yang G, Larocque A, Régina A, Demeule M, Sarret P, Gendron L. Use of a Noninvasive Brain-Penetrating Peptide-Drug Conjugate Strategy to Improve the Delivery of Opioid Pain Relief Medications to the Brain. J Pharmacol Exp Ther 2020; 374:52-61. [DOI: 10.1124/jpet.119.263566] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Indexed: 02/04/2023] Open
|
205
|
Cao Z, Yan L, Shen Z, Chen Y, Shi Y, He X, Zhou N. A novel splice variant of Gαq-coupled Bombyx CAPA-PVK receptor 1 functions as a specific Gαi/o-linked receptor for CAPA-PK. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118718. [PMID: 32289337 DOI: 10.1016/j.bbamcr.2020.118718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 01/04/2023]
Abstract
Alternative splicing enables G protein-coupled receptor (GPCR) genes to greatly increase the number of structurally and functionally distinct receptor isoforms. However, the functional role and relevance of the individual GPCR splice variants in regulating physiological processes are still to be assessed. A naturally occurring alternative splice variant of Bombyx CAPA-PVK receptor, BomCAPA-PVK-R1-Δ341, has been shown to act as a dominant-negative protein to regulate cell surface expression and function of the canonical CAPA-PVK receptor. Herein, using functional assays, we identify the splice variant Δ341 as a specific receptor for neuropeptide CAPA-PK, and upon activation, Δ341 signals to ERK1/2 pathway. Further characterization demonstrates that Δ341 couples to Gαi/o, distinct from the Gαq-coupled canonical CAPA-PVK receptor, triggering ERK1/2 phosphorylation through Gβγ-PI3K-PKCζ signaling cascade. Moreover, our ELISA data show that the ligand-dependent internalization of the splice variant Δ341 is significantly impaired due to lack of GRKs-mediated phosphorylation sites. Our findings highlight the potential of this knowledge for molecular, pharmacological and physiological studies on GPCR splice variants in the future.
Collapse
Affiliation(s)
- Zheng Cao
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lili Yan
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhangfei Shen
- Department of Economic Zoology, College of Animal Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yu Chen
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ying Shi
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaobai He
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
206
|
Sohrevardi SM, Pournamdari M, Salimi R, Sarrafzadeh F, Ahmadinejad M. Comparing the Efficacy of Methadone and Tincture of Opium in Controlling Agitation Caused by Withdrawal Syndrome in Opium-Addicted Patients in the Intensive Care Unit: A Randomized Trial Study. ADDICTION & HEALTH 2020; 12:69-76. [PMID: 32782729 PMCID: PMC7395929 DOI: 10.22122/ahj.v12i2.259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/18/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Few studies have been conducted regarding the comparison of the efficacy of methadone and tincture of opium (TOP) in controlling agitation induced by withdrawal syndrome. Therefore, the current randomized trial study is carried out with the aim to evaluate comparisons on the efficacy of methadone and TOP in controlling agitation caused by withdrawal syndrome in opium addicted patients in the intensive care units (ICUs). METHODS This clinical trial study was conducted on 60 patients admitted to ICU of Shahid Bahonar Hospital, Kerman, Iran. After classification of the patients into two groups, the first and second groups consumed methadone syrup (5 mg/ml) and TOP (10 mg/ml), respectively. Agitation in these patients was assessed through the Richmond Agitation-Sedation Scale (RASS). Vital signs were also assessed. Paired sample t-test and independent t-test were used for data analysis. FINDINGS In the current study, the administered dose of methadone and TOP was 36.17 ± 26.99 and 112.67 ± 102.74 mg, respectively (P < 0.010). Methadone administration led to a significant decrease of the patients' vital signs, including systolic blood pressure, heart rate, respiratory rate, and Glasgow Coma Scale (GCS) (P < 0.05). Though TOP administration decreased systolic blood pressure and GCS significantly (P < 0.05), it had no effect on patients' diastolic blood pressure, body temperature, heart rate, and respiratory rate (P > 0.05). In total, no significant difference was detected between two groups regarding vital signs (P > 0.05). However, a significant difference was seen between methadone and TOP groups in terms of RASS score (P < 0.01). CONCLUSION According to the results of the current study, lower dose of methadone, compared to TOP, could control agitation caused by opium withdrawal symptoms.
Collapse
Affiliation(s)
- Seyed Mojtaba Sohrevardi
- Robarts Research Institute, University of Western Ontario, Ontario, Canada AND Pharmaceutical Sciences Research Center AND Department of Clinical Pharmacy, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mostafa Pournamdari
- Pharmaceutics Research Center AND Department of Medical Chemistry, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Razieh Salimi
- Pharmaceutics Research Center AND Department of Medical Chemistry, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Farhad Sarrafzadeh
- Department of Internal Medicine, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Ahmadinejad
- Department of Anesthesia, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
207
|
Yadav SK, Nagar DP, Bhattacharya R. Effect of fentanyl and its three novel analogues on biochemical, oxidative, histological, and neuroadaptive markers after sub-acute exposure in mice. Life Sci 2020; 246:117400. [PMID: 32032645 DOI: 10.1016/j.lfs.2020.117400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/19/2020] [Accepted: 02/02/2020] [Indexed: 10/25/2022]
Abstract
AIMS Comparative sub-acute toxicity, including tolerance and dependence potential of fentanyl and its three novel and potent analogues was determined in mice. MAIN METHODS Comparative sub-acute (21 d, intraperitoneal; 1/10 LD50) toxicity of fentanyl and its three novel analogues viz., N-(1-(2-phenoxyethyl)-4-piperidinyl) propionanilide (2), N-isopropyl-3-(4-(N-phenylpropionamido)piperidin-1-yl)propanamide (5), and N-t-butyl-3-(4-(N-phenylpropionamido)piperidin-1-yl)propanamide (6) was determined in mice. Animals were observed for additional seven days to assess the recovery. The brain, liver and kidney toxicity was assessed on the basis of various biochemical, oxidative, histological, and neuroadaptive markers. The expression levels of key neuronal markers associated with drug tolerance and dependence were investigated by western blot and immunohistochemistry. KEY FINDINGS Fentanyl and its analogues caused abnormal levels of liver and kidney specific biomarkers in plasma. Brain malondialdehyde (MDA) levels were raised by all the treatments and kidney MDA level by analogue 6 (21 d). Reduced glutathione levels in brain, liver, and kidney were diminished by all the treatments (21 & 28 d) and a significant change in the levels of antioxidant enzymes was also produced mainly after 21 d. The deleterious effects of fentanyl and its analogues were further substantiated by corresponding histopathological changes in brain, liver and kidney (21 & 28 d). These compounds were also found to produce neuroadaptive changes as evidenced by the increased expression levels of c-Fos, glucocorticoid receptor, N-methyl-d-aspartate receptor1 and μ-opioid receptor (21 & 28 d). SIGNIFICANCE Three novel analogues of fentanyl were envisaged to have alternative therapeutic potentials. However, their comparative sub-acute toxicity revealed undesirable side effects, thereby masking their therapeutic ability.
Collapse
Affiliation(s)
- Shiv Kumar Yadav
- Division of Pharmacology and Toxicology, Defence Research and Development Establishment, Jhansi Road, Gwalior 474 002, M.P., India
| | - D P Nagar
- Division of Pharmacology and Toxicology, Defence Research and Development Establishment, Jhansi Road, Gwalior 474 002, M.P., India
| | - Rahul Bhattacharya
- Division of Pharmacology and Toxicology, Defence Research and Development Establishment, Jhansi Road, Gwalior 474 002, M.P., India.
| |
Collapse
|
208
|
N-Phenethyl Substitution in 14-Methoxy-N-methylmorphinan-6-ones Turns Selective µ Opioid Receptor Ligands into Dual µ/δ Opioid Receptor Agonists. Sci Rep 2020; 10:5653. [PMID: 32221355 PMCID: PMC7101422 DOI: 10.1038/s41598-020-62530-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/21/2020] [Indexed: 11/12/2022] Open
Abstract
Morphine and structurally-derived compounds are µ opioid receptor (µOR) agonists, and the most effective analgesic drugs. However, their usefulness is limited by serious side effects, including dependence and abuse potential. The N-substituent in morphinans plays an important role in opioid activities in vitro and in vivo. This study presents the synthesis and pharmacological evaluation of new N-phenethyl substituted 14-O-methylmorphinan-6-ones. Whereas substitution of the N-methyl substituent in morphine (1) and oxymorphone (2) by an N-phenethyl group enhances binding affinity, selectivity and agonist potency at the µOR of 1a and 2a, the N-phenethyl substitution in 14-methoxy-N-methylmorphinan-6-ones (3 and 4) converts selective µOR ligands into dual µ/δOR agonists (3a and 4a). Contrary to N-methylmorphinans 1–4, the N-phenethyl substituted morphinans 1a–4a produce effective and potent antinociception without motor impairment in mice. Using docking and molecular dynamics simulations with the µOR, we establish that N-methylmorphinans 1–4 and their N-phenethyl counterparts 1a–4a share several essential receptor-ligand interactions, but also interaction pattern differences related to specific structural features, thus providing a structural basis for their pharmacological profiles. The emerged structure-activity relationships in this class of morphinans provide important information for tuning in vitro and in vivo opioid activities towards discovery of effective and safer analgesics.
Collapse
|
209
|
De Caro C, Raucci F, Saviano A, Cristiano C, Casillo GM, Di Lorenzo R, Sacchi A, Laneri S, Dini I, De Vita S, Chini MG, Bifulco G, Calignano A, Maione F, Mascolo N. Pharmacological and molecular docking assessment of cryptotanshinone as natural-derived analgesic compound. Biomed Pharmacother 2020; 126:110042. [PMID: 32203893 DOI: 10.1016/j.biopha.2020.110042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 11/19/2022] Open
Abstract
Medicinal plants from traditional chinese medicine are used increasingly worldwide for their benefits to health and quality of life for the relevant clinical symptoms related to pain. Among them, Salvia miltiorrhiza Bunge is traditionally used in asian countries as antioxidant, anticancer, anti-inflammatory and analgesic agent. In this context, several evidences support the hypothesis that some tanshinones, in particular cryptotanshinone (CRY), extracted from the roots (Danshen) of this plant exhibit analgesic actions. However, it is surprisingly noted that no pharmacological studies have been carried out to explore the possible analgesic action of this compound in terms of modulation of peripheral and/or central pain. Therefore, in the present study, by using peripheral and central pain models of nociception, such as tail flick and hot plate test, the analgesic effect of CRY in mice was evaluated. Successively, by the aim of a computational approach, we have evaluated the interaction mode of this diterpenoid on opioid and cannabinoid system. Finally, CRY was dosed in mice serum by an HPLC method validated according to European Medicines Agency guidelines validation rules. Here, we report that CRY displayed anti-nociceptive activity on both hot plate and tail flick test, with a prominent long-lasting peripheral analgesic effect. These evidences were indirectly confirmed after the daily administration of the tanshinone for 7 and 14 days. In addition, the analgesic effect of CRY was reverted by naloxone and cannabinoid antagonists and amplified by arginine administration. These findings were finally supported by HPLC and docking studies, that revealed a noteworthy presence of CRY on mice serum 1 h after its intraperitoneal administration and a possible interaction of tested compound on μ and k receptors. Taken together, these results provide a new line of evidences showing that CRY can produce analgesia against various phenotypes of nociception with a mechanism that seems to be related to an agonistic activity on opioid system.
Collapse
Affiliation(s)
- Carmen De Caro
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Anella Saviano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Claudia Cristiano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Ritamaria Di Lorenzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Antonia Sacchi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Sonia Laneri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Irene Dini
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Simona De Vita
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, Salerno, Italy
| | - Maria Giovanna Chini
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, Salerno, Italy; Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, Isernia, I-86090, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, Salerno, Italy.
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy.
| | - Nicola Mascolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| |
Collapse
|
210
|
Assessment of structure-activity relationships and biased agonism at the Mu opioid receptor of novel synthetic opioids using a novel, stable bio-assay platform. Biochem Pharmacol 2020; 177:113910. [PMID: 32179045 DOI: 10.1016/j.bcp.2020.113910] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 03/10/2020] [Indexed: 11/20/2022]
Abstract
Fentanyl and morphine are agonists of the Mu opioid receptor (MOR), which is a member of the GPCR family. Their analgesic effects are associated with unwanted side effects. On a signaling level downstream from MOR, it has been hypothesized that analgesia may be mediated through the G protein pathway, whereas the undesirable effects of opioids have been linked to the β-arrestin (βarr) pathway. Despite being an increasingly debated subject, little is known about a potential 'bias' (i.e. the preferential activation of one pathway over the other) of the novel synthetic opioids (NSO) - including fentanyl analogs - that have emerged on the illegal drug market. We have therefore developed and applied a novel, robust bio-assay platform to study the activity of 21 NSO, to evaluate to what extent these MOR agonists show biased agonism and to investigate the potential correlation with their structure. In addition, we evaluated the functional selectivity of TRV130, a purported G protein-biased agonist. We applied newly established stable bio-assays in HEK293T cells, based on the principle of functional complementation of a split nanoluciferase, to assess MOR activation via recruitment of a mini-Gi protein (GTPase domain of Gαi subunit) or βarr2. All but two of the tested NSO demonstrated a concentration-dependent response at MOR in both bio-assays. The developed bio-assays allow to gain insight into the βarr2 or G protein recruitment potential of NSO, which may eventually help to better understand why certain opioids are associated with higher toxicity. Adding to the recent discussion about the relevance of the biased agonism concept for opioids, we did not observe a significant bias for any of the evaluated compounds, including TRV130.
Collapse
|
211
|
Busserolles J, Lolignier S, Kerckhove N, Bertin C, Authier N, Eschalier A. Replacement of current opioid drugs focusing on MOR-related strategies. Pharmacol Ther 2020; 210:107519. [PMID: 32165137 DOI: 10.1016/j.pharmthera.2020.107519] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
The scarcity and limited risk/benefit ratio of painkillers available on the market, in addition to the opioid crisis, warrant reflection on new innovation strategies. The pharmacopoeia of analgesics is based on products that are often old and derived from clinical empiricism, with limited efficacy or spectrum of action, or resulting in an unsatisfactory tolerability profile. Although they are reference analgesics for nociceptive pain, opioids are subject to the same criticism. The use of opium as an analgesic is historical. Morphine was synthesized at the beginning of the 19th century. The efficacy of opioids is limited in certain painful contexts and these drugs can induce potentially serious and fatal adverse effects. The current North American opioid crisis, with an ever-rising number of deaths by opioid overdose, is a tragic illustration of this. It is therefore legitimate to develop research into molecules likely to maintain or increase opioid efficacy while improving their tolerability. Several avenues are being explored including targeting of the mu opioid receptor (MOR) splice variants, developing biased agonists or targeting of other receptors such as heteromers with MOR. Ion channels acting as MOR effectors, are also targeted in order to offer compounds without MOR-dependent adverse effects. Another route is to develop opioid analgesics with peripheral action or limited central nervous system (CNS) access. Finally, endogenous opioids used as drugs or compounds that modify the metabolism of endogenous opioids (Dual ENKephalinase Inhibitors) are being developed. The aim of the present review is to present these various targets/strategies with reference to current indications for opioids, concerns about their widespread use, particularly in chronic non-cancer pains, and ways of limiting the risk of opioid abuse and misuse.
Collapse
Affiliation(s)
- Jérôme Busserolles
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Stéphane Lolignier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Nicolas Kerckhove
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Célian Bertin
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Nicolas Authier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
212
|
Kvello AMS, Andersen JM, Boix F, Mørland J, Bogen IL. The role of 6-acetylmorphine in heroin-induced reward and locomotor sensitization in mice. Addict Biol 2020; 25:e12727. [PMID: 30788879 DOI: 10.1111/adb.12727] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 01/11/2023]
Abstract
We have previously demonstrated that heroin's first metabolite, 6-acetylmorphine (6-AM), is an important mediator of heroin's acute effects. However, the significance of 6-AM to the rewarding properties of heroin still remains unknown. The present study therefore aimed to examine the contribution of 6-AM to heroin-induced reward and locomotor sensitization. Mice were tested for conditioned place preference (CPP) induced by equimolar doses of heroin or 6-AM (1.25-5 μmol/kg). Psychomotor activity was recorded during the CPP conditioning sessions for assessment of drug-induced locomotor sensitization. The contribution of 6-AM to heroin reward and locomotor sensitization was further examined by pretreating mice with a 6-AM specific antibody (anti-6-AM mAb) 24 hours prior to the CPP procedure. Both heroin and 6-AM induced CPP in mice, but heroin generated twice as high CPP scores compared with 6-AM. Locomotor sensitization was expressed after repeated exposure to 2.5 and 5 μmol/kg heroin or 6-AM, but not after 1.25 μmol/kg, and we found no correlation between the expression of CPP and the magnitude of locomotor sensitization for either opioid. Pretreatment with anti-6-AM mAb suppressed both heroin-induced and 6-AM-induced CPP and locomotor sensitization. These findings provide evidence that 6-AM is essential for the rewarding and sensitizing properties of heroin; however, heroin caused stronger reward compared with 6-AM. This may be explained by the higher lipophilicity of heroin, providing more efficient drug transfer to the brain, ensuring rapid increase in the brain 6-AM concentration.
Collapse
Affiliation(s)
- Anne Marte Sjursen Kvello
- Section for Drug Abuse Research, Department of Forensic SciencesOslo University Hospital Oslo Norway
- School of Pharmacy, Faculty of Mathematics and Natural SciencesUniversity of Oslo Oslo Norway
| | - Jannike Mørch Andersen
- Section for Drug Abuse Research, Department of Forensic SciencesOslo University Hospital Oslo Norway
- School of Pharmacy, Faculty of Mathematics and Natural SciencesUniversity of Oslo Oslo Norway
| | - Fernando Boix
- Section for Drug Abuse Research, Department of Forensic SciencesOslo University Hospital Oslo Norway
| | - Jørg Mørland
- Division of Health Data and DigitalisationNorwegian Institute of Public Health Oslo Norway
| | - Inger Lise Bogen
- Section for Drug Abuse Research, Department of Forensic SciencesOslo University Hospital Oslo Norway
- Institute of Basic Medical Sciences, Faculty of MedicineUniversity of Oslo Oslo Norway
| |
Collapse
|
213
|
The effect of endurance exercise and methadone on μ-opioid receptor gene expression in morphine-dependent rats following withdrawal syndrome. SPORT SCIENCES FOR HEALTH 2020. [DOI: 10.1007/s11332-019-00596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
214
|
Oesterle TS, Kolla BP, Rummans TA, Gold MS. Medication-assisted therapies for opioid use disorders in patients with chronic pain. J Neurol Sci 2020; 411:116728. [PMID: 32092625 DOI: 10.1016/j.jns.2020.116728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/15/2020] [Accepted: 02/06/2020] [Indexed: 01/30/2023]
Abstract
Opioids have been used to treat pain and invoke pleasure for centuries. Modern scientific advancements have led to more potent, synthetic opioids. While certainly more effective in treating pain, they can also be much more addictive. Over the years the scientific community has developed a clearer understanding of the role opioid receptors play in causing and treating opioid use disorders (OUD) and we now know that OUD can develop in individuals taking opioids for "legitimate" pain. Current guidelines suggest that all prescribers (especially those prescribing opioids) be capable treating OUD. Pharmacological advances have led to a wide array of safe and effective treatment options to address OUDs. This paper will discuss the history of opioid development, what is known about the transition from analgesic uses to addiction and modern evidenced based treatment strategies to address OUDs.
Collapse
Affiliation(s)
- Tyler S Oesterle
- Mayo Clinic - Rochester, Department of Psychiatry & Psychology, 200 First Street SW, Rochester, MN 55905, United States of America.
| | - Bhanu Prakash Kolla
- Mayo Clinic - Rochester, Department of Psychiatry & Psychology, 200 First Street SW, Rochester, MN 55905, United States of America
| | - Teresa A Rummans
- Mayo Clinic - Rochester, Department of Psychiatry & Psychology, 200 First Street SW, Rochester, MN 55905, United States of America
| | - Mark S Gold
- Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| |
Collapse
|
215
|
Summer A, Di Frangia F, Ajmone Marsan P, De Noni I, Malacarne M. Occurrence, biological properties and potential effects on human health of β-casomorphin 7: Current knowledge and concerns. Crit Rev Food Sci Nutr 2020; 60:3705-3723. [PMID: 32033519 DOI: 10.1080/10408398.2019.1707157] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The genetic variant A1 of bovine β-casein (β-Cn) presents a His residue at a position 67 of the mature protein. This feature makes the Ile66-His67 bond more vulnerable to enzymatic cleavage, determining the release of the peptide β-Cn f(60-66), named β-casomorphin 7 (BCM7). BCM7 is an opioid-agonist for μ receptors, and it has been hypothesized to be involved in the development of different non-transmissible diseases in humans. In the last decade, studies have provided additional results on the potential health impact of β-Cn A1 and BCM7. These studies, here reviewed, highlighted a relation between the consumption of β-Cn A1 (and its derivative BCM7) and the increase of inflammatory response as well as discomfort at the gastrointestinal level. Conversely, the role of BCM7 and the effects of ingestion of β-Cn A1 on the onset or worsening of other non-transmissible diseases as caused or favored by still need proof of evidence. Overall, the reviewed literature demonstrates that the "β-Cn A1/BCM7 issue" remains an intriguing but not exhaustively explained topic in human nutrition. On this basis, policies in favor of breeding for β-Cn variants not releasing BCM7 and consumption of "A1-like" milk appear not yet sound for a healthier and safer nutrition.
Collapse
Affiliation(s)
- Andrea Summer
- Department of Veterinary Sciences, Università di Parma, Parma, Italy
| | | | - Paolo Ajmone Marsan
- Department of Animal Science, Food and Nutrition (DIANA) and Nutrigenomics and Proteomics Research Center (PRONUTRIGEN), Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Ivano De Noni
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Massimo Malacarne
- Department of Veterinary Sciences, Università di Parma, Parma, Italy
| |
Collapse
|
216
|
Levran O, Even-Tov E, Zhao L. A hominid-specific shift in cerebellar expression, upstream retrotransposons, and a potential cis-regulatory mechanism: bioinformatics analyses of the mu-opioid receptor gene. Heredity (Edinb) 2020; 124:325-335. [PMID: 31712748 PMCID: PMC6972845 DOI: 10.1038/s41437-019-0282-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/26/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
The mu-opioid receptors (MOR, OPRM1) mediate the effects of beta-endorphin and modulate many biological functions including reward processing and addiction. The present study aimed to use bioinformatics to determine OPRM1 brain expression profiles in higher primates and to look for regulatory mechanisms. We used the same computational pipeline to analyze publicly available expression data from postmortem brain regions across humans, chimpanzees, and rhesus macaques. The most intriguing finding was high OPRM1 cerebellar expression in humans and chimpanzees and low expression in macaques. Together with previous reports of low cerebellar OPRM1 expression in mice, this suggests an evolutionary shift in the expression profiles. Bioinformatic analysis of the OPRM1 upstream region revealed a functional CTCF-binding region that evolved from tandem insertions of retrotransposons L1P1 and L1PA1 upstream (-60 kb) of OPRM1. The insertions arose in different time points after the split of small apes from great apes, and their combined sequence is unique. Furthermore, the derived G allele of SNP rs12191876, in the inserted region, is associated with an increased OPRM1 expression in the cerebellum of postmortem human brains (p = 4.7e-5). The derived G allele became the major allele (60-90%) in the populations represented in the 1000 Genomes Project and may be beneficial. This study provides a foundation for building new knowledge about evolutionary differences in OPRM1 brain expression. Further investigations are needed to elucidate the role of the inserted region and its SNPs in OPRM1 expression, and to assess the biological function and relevance of OPRM1 expression in the cerebellum.
Collapse
Affiliation(s)
- Orna Levran
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA.
| | - Eran Even-Tov
- Laboratory of Evolutionary Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Li Zhao
- Laboratory of Evolutionary Genetics and Genomics, The Rockefeller University, New York, NY, USA
| |
Collapse
|
217
|
Lu Z, Xu J, Wang Q, Pan YX. Morphine modulates the expression of mu-opioid receptor exon 5-associated full-length C-terminal splice variants by upregulating miR-378a-3p. FASEB J 2020; 34:4540-4556. [PMID: 31999011 DOI: 10.1096/fj.201901879rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 11/11/2022]
Abstract
The mu-opioid receptor gene, OPRM1, undergoes extensive alternative splicing, creating an array of splice variants that are conserved from rodent to human. Both mouse and human OPRM1 have five exon 5-associated seven transmembrane full-length carboxyl terminal variants, MOR-1B1, MOR-1B2, MOR-1B3, MOR-1B4, and MOR-1B5, all of which are derived from alternative 3' splicing from exon 3 to alternative sites within exon 5. The functional relevance of these exon 5-associated MOR-1Bs has been demonstrated in mu agonist-induced G protein coupling, adenylyl cyclase activity, receptor internalization and desensitization, and post-endocytic sorting, as well as region-specific expression at the mRNA level. In the present study, we mapped a polyadenylation site for both mouse and human MOR-1Bs that defines the 3'-untranslated regions (3'-UTR) of MOR-1Bs and stabilizes mMOR-1Bs mRNAs. We identified a conserved miR378a-3p sequence in the 3'-UTR of both mouse and human MOR-1BS transcripts through which miR-378a-3p can regulate the expression of MOR-1Bs at the mRNA level. Chronic morphine treatment significantly increased the miR-378-3p level in Be(2)C cells and the brainstem of the morphine tolerant mice, contributing to the decreased expression of the mouse and human MOR-1B3 and MOR-1B4. Our study provides new insights into the role of miRNAs and Oprm1 splice variants in morphine tolerance.
Collapse
Affiliation(s)
- Zhigang Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,The Affiliated Hospital of Nanjing University of Chinese Medicine, First College of Clinical Medicine, Nanjing, China.,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin Xu
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Qian Wang
- International Education College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying-Xian Pan
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Program in Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
218
|
Maraschin JC, Sestile CC, Yabiku CT, Roncon CM, de Souza Fiaes GC, Graeff FG, Audi EA, Zangrossi H. Effects of the adjunctive treatment of antidepressants with opiorphin on a panic-like defensive response in rats. Behav Brain Res 2020; 378:112263. [PMID: 31568834 DOI: 10.1016/j.bbr.2019.112263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Antidepressants are the first-choice for pharmacological treatment of panic disorder. However, they present disadvantages, such as delayed therapeutic effect, many side effects and a considerable rate of non-responders. These shortcomings prompt the development of new therapeutic strategies. Among these are the adjunctive use of enkephalinase inhibitors, such as opiorphin, which supposedly acts by increasing the availability of brain enkephalins and other endogenous opioids. AIMS We here evaluated whether opiorphin in the dorsal periaqueductal grey matter (dPAG), a key panic-related area, accelerates and/or facilitates the antipanic-like effect of fluoxetine or imipramine. We also verified whether the panicolytic effect of imipramine depends on activation of μ-opioid receptors (MORs). METHODS Male Wistar rats were submitted to the escape task of the elevated T-maze, an index of panic attack, after treatment with imipramine (3, 7 or 21 days) or fluoxetine (3, 7, 14 or 21 days), combined with an intra-dPAG injection of opiorphin. RESULTS Opiorphin facilitated and accelerated the panicolytic-like effect caused by imipramine, but not with fluoxetine. The antipanic-like effect caused by chronic imipramine did not depend on MOR activation in the dPAG. CONCLUSION Combined treatment of antidepressant drugs with opiorphin for hastening or potentiating the effects of the former compounds may not be generally effective, with the results varying depending on the type/class of these panicolytic drugs.
Collapse
Affiliation(s)
- Jhonatan Christian Maraschin
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, Maringá, Paraná, 87020-900, Brazil
| | - Caio César Sestile
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, Maringá, Paraná, 87020-900, Brazil
| | - Cláudia Tiemi Yabiku
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, Maringá, Paraná, 87020-900, Brazil
| | - Camila Marroni Roncon
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, Maringá, Paraná, 87020-900, Brazil
| | - Gislaine Cardoso de Souza Fiaes
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, Maringá, Paraná, 87020-900, Brazil
| | - Frederico Guilherme Graeff
- Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo, 14050-220, Brazil; Neurobiology of Emotions Research Centre (NAP-USP-NuPNE), School of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Av. dos Bandeirantes, 3900, Ribeirao Preto, São Paulo, 14049-900, Brazil
| | - Elisabeth Aparecida Audi
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, Maringá, Paraná, 87020-900, Brazil; Behavioural Neurosciences Institute (INeC), Av. do Café, 2450, Monte Alegre, Ribeirão Preto, São Paulo, 14050-220, Brazil; Neurobiology of Emotions Research Centre (NAP-USP-NuPNE), School of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Av. dos Bandeirantes, 3900, Ribeirao Preto, São Paulo, 14049-900, Brazil
| | - Hélio Zangrossi
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Av. dos Bandeirantes, 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil.
| |
Collapse
|
219
|
Aversion No MOR: Mu-opioid receptors in habenular β4 neurons are key for naloxone aversion. Neuropsychopharmacology 2020; 45:243-244. [PMID: 31055593 PMCID: PMC6901489 DOI: 10.1038/s41386-019-0404-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 11/08/2022]
|
220
|
Puryear CB, Brooks J, Tan L, Smith K, Li Y, Cunningham J, Todtenkopf MS, Dean RL, Sanchez C. Opioid receptor modulation of neural circuits in depression: What can be learned from preclinical data? Neurosci Biobehav Rev 2020; 108:658-678. [DOI: 10.1016/j.neubiorev.2019.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
|
221
|
Opioid treatment for acute and chronic pain in patients with sickle cell disease. Neurosci Lett 2020; 714:134534. [DOI: 10.1016/j.neulet.2019.134534] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 09/13/2019] [Accepted: 10/01/2019] [Indexed: 12/24/2022]
|
222
|
Molecular Basis of Opioid Action: From Structures to New Leads. Biol Psychiatry 2020; 87:6-14. [PMID: 31653480 PMCID: PMC6898784 DOI: 10.1016/j.biopsych.2019.08.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/30/2019] [Accepted: 08/31/2019] [Indexed: 02/06/2023]
Abstract
Since the isolation of morphine from the opium poppy over 200 years ago, the molecular basis of opioid action has remained the subject of intense inquiry. The identification of specific receptors responsible for opioid function and the discovery of many chemically diverse molecules with unique opioid-like efficacies have provided glimpses into the molecular logic of opioid action. Recent revolutions in the structural biology of transmembrane proteins have, for the first time, yielded high-resolution views into the 3-dimensional shapes of all 4 opioid receptors. These studies have begun to decode the chemical logic that enables opioids to specifically bind and activate their receptor targets. A combination of spectroscopic experiments and computational simulations has provided a view into the molecular movements of the opioid receptors, which itself gives rise to the complex opioid pharmacology observed at the cellular and behavioral levels. Further diversity in opioid receptor structure is driven by both genetic variation and receptor oligomerization. These insights have enabled computational drug discovery efforts, with some evidence of success in the design of completely novel opioids with unique efficacies. The combined progress over the past few years provides hope for new, efficacious opioids devoid of the side effects that have made them the scourge of humanity for millennia.
Collapse
|
223
|
Morgan MM, Tran A, Wescom RL, Bobeck EN. Differences in antinociceptive signalling mechanisms following morphine and fentanyl microinjections into the rat periaqueductal gray. Eur J Pain 2019; 24:617-624. [PMID: 31785128 DOI: 10.1002/ejp.1513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/08/2019] [Accepted: 11/25/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Morphine and fentanyl are two of the most commonly used opioids to treat pain. Although both opioids produce antinociception by binding to mu-opioid receptors (MOR), they appear to act via distinct signalling pathways. OBJECTIVE This study will reveal whether differences in morphine and fentanyl antinociception are the result of selective activation of G-protein signalling and/or selective activation of pre- or postsynaptic MORs. METHODS The contribution of each mechanism to morphine and fentanyl antinociception was assessed by microinjecting drugs to alter G-protein signalling or block potassium channels linked to pre- and postsynaptic MORs in the ventrolateral periaqueductal gray (PAG) of male Sprague-Dawley rats. RESULTS Both morphine and fentanyl produced a dose-dependent antinociception when microinjected into the PAG. Enhancement of intracellular G-protein signalling by microinjection of the Regulator of G-protein Signalling 4 antagonist CCG-63802 into the PAG enhanced the antinociceptive potency of morphine, but not fentanyl. Microinjection of α-dendrotoxin into the PAG to block MOR activation of presynaptic Kv + channels caused a significant rightward shift in the dose-response curve of both morphine and fentanyl. Microinjection of tertiapin-Q to block MOR activation of postsynaptic GIRK channels caused a larger shift in the dose-response curve for fentanyl than morphine antinociception. CONCLUSIONS These findings reveal different PAG signalling mechanisms for morphine and fentanyl antinociception. In contrast with fentanyl, the antinociceptive effects of morphine are mediated by G-protein signalling primarily activated by presynaptic MORs. SIGNIFICANCE Microinjection of the opioids morphine and fentanyl into the periaqueductal gray (PAG) produce antinociception via mu-opioid receptor signalling. This study reveals differences in the signalling mechanisms underlying morphine and fentanyl antinociception in the PAG. In contrast with fentanyl, morphine antinociception is primarily mediated by presynaptic opioid receptors and is enhanced by blocking RGS proteins.
Collapse
Affiliation(s)
- Michael M Morgan
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, USA
| | - Alexander Tran
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, USA
| | - Rebecca L Wescom
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, USA
| | - Erin N Bobeck
- Department of Biology, Utah State University, Logan, UT, USA
| |
Collapse
|
224
|
Xiao L, Wang Y, Zhang M, Wu W, Kong L, Ma Y, Xu X, Liu X, He Q, Qian Y, Sun H, Wu H, Lin C, Huang H, Ye R, Jiang S, Ye RF, Yuan C, Fang S, Xue D, Yang X, Chen H, Zheng Y, Yu L, Xie Q, Zheng L, Fu W, Li W, Qiu Z, Liu J, Shao L. Discovery of a Highly Selective and Potent κ Opioid Receptor Agonist from N-Cyclopropylmethyl-7α-phenyl-6,14-endoethanotetrahydronorthebaines with Reduced Central Nervous System (CNS) Side Effects Navigated by the Message-Address Concept. J Med Chem 2019; 62:11054-11070. [PMID: 31738550 DOI: 10.1021/acs.jmedchem.9b00857] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Effective and safe analgesics represent an unmet medical need for the treatment of acute and chronic pain. A series of N-cyclopropylmethyl-7α-phenyl-6,14-endoethanotetrahydronorthebaines were designed, synthesized, and assayed, leading to the discovery of a benzylamine derivative (compound 4, SLL-039) as a highly selective and potent κ opioid agonist (κ, Ki = 0.47 nM, κ/μ = 682, κ/δ = 283), which was confirmed by functional assays in vitro and antinociceptive assays in vivo. The in vivo effect could be blocked by pretreatment with the selective κ antagonist nor-BNI. Moreover, this compound did not induce sedation, a common dose limiting effect of κ opioid receptor agonists, at its analgesic dose compared to U50,488H. The dissociation of sedation/antinociception found in SLL-039 was assumed to be correlated with the occupation of its benzamide motif in a unique subsite involving V1182.63, W124EL1, and E209EL2.
Collapse
Affiliation(s)
- Li Xiao
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Yujun Wang
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Mumei Zhang
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Weiwei Wu
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China.,University of Chinese Academy of Sciences , No. 19A Yuquan Road , Beijing 100049 , China
| | - Linghui Kong
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Yan Ma
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China.,Shanghai University School of Life Sciences , No. 99 Shangda Road , Shanghai 200444 , China
| | - Xuejun Xu
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Xiao Liu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Qian He
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Yuanyuan Qian
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Huijiao Sun
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Haihao Wu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Cheng Lin
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Huoming Huang
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Rongrong Ye
- Shanghai Institute of Technology , No. 100 Haiquan Road , Shanghai 201418 , China
| | - Shuang Jiang
- Nanjing University of Chinese Medicine , No. 138 Xianlin Avenue , Nanjing 210023 , China
| | - Ru-Feng Ye
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Congmin Yuan
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Shengyang Fang
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Dengqi Xue
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Xicheng Yang
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Hao Chen
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Yilin Zheng
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Linqian Yu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Lan Zheng
- Minhang Hospital , Fudan University , No. 170 Xinsong Road , Shanghai 201199 , China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Zhuibai Qiu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Jinggen Liu
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China.,University of Chinese Academy of Sciences , No. 19A Yuquan Road , Beijing 100049 , China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China.,State Key Laboratory of Medical Neurobiology , Fudan University , No. 138 Yixueyuan Road , Shanghai 200032 , China
| |
Collapse
|
225
|
Mata X, Renaud G, Mollereau C. The repertoire of family A-peptide GPCRs in archaic hominins. Peptides 2019; 122:170154. [PMID: 31560950 DOI: 10.1016/j.peptides.2019.170154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/30/2022]
Abstract
Given the importance of G-protein coupled receptors in the regulation of many physiological functions, deciphering the relationships between genotype and phenotype in past and present hominin GPCRs is of main interest to understand the evolutionary process that contributed to the present-day variability in human traits and health. Here, we carefully examined the publicly available genomic and protein sequence databases of the archaic hominins (Neanderthal and Denisova) to draw up the catalog of coding variations in GPCRs for peptide ligands, in comparison with living humans. We then searched in the literature the functional changes, phenotypes and risk of disease possibly associated with the detected variants. Our survey suggests that Neanderthal and Denisovan hominins were likely prone to lower risk of obesity, to enhanced platelet aggregation in response to thrombin, to better response to infection, to less anxiety and aggressiveness and to favorable sociability. While some archaic variants were likely advantageous in the past, they might be responsible for maladaptive disorders today in the context of modern life and/or specific regional distribution. For example, an archaic haplotype in the neuromedin receptor 2 is susceptible to confer risk of diabetic nephropathy in type 1 diabetes in present-day Europeans. Paying attention to the pharmacological properties of some of the archaic variants described in this study may be helpful to understand the variability of therapeutic efficacy between individuals or ethnic groups.
Collapse
Affiliation(s)
- Xavier Mata
- Laboratoire Anthropologie Moléculaire et Imagerie de Synthèse (AMIS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Gabriel Renaud
- Centre for GeoGenetics, Natural History Museum of Denmark, University of Copenhagen, Copenhagen K, Denmark
| | - Catherine Mollereau
- Laboratoire Anthropologie Moléculaire et Imagerie de Synthèse (AMIS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
226
|
µ-opioid receptor-mediated downregulation of midline thalamic pathways to basal and central amygdala. Sci Rep 2019; 9:17837. [PMID: 31780740 PMCID: PMC6882837 DOI: 10.1038/s41598-019-54128-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 11/04/2019] [Indexed: 11/08/2022] Open
Abstract
Brain µ-opioid receptors (MOR) mediate reward and help coping with pain, social rejection, anxiety and depression. The dorsal midline thalamus (dMT) integrates visceral/emotional signals and biases behavior towards aversive or defensive states through projections to the amygdala. While a dense MOR expression in the dMT has been described, the exact cellular and synaptic mechanisms of µ-opioidergic modulation in the dMT-amygdala circuitry remain unresolved. Here, we hypothesized that MORs are important negative modulators of dMT-amygdala excitatory networks. Using retrograde tracers and targeted channelrhodopsin expression in combination with patch-clamp electrophysiology, we found that projections of dMT neurons onto both basal amygdala principal neurons (BA PN) and central amygdala (CeL) neurons are attenuated by stimulation of somatic or synaptic MORs. Importantly, dMT efferents to the amygdala drive feedforward excitation of centromedial amygdala neurons (CeM), which is dampened by MOR activation. This downregulation of excitatory activity in dMT-amygdala networks puts the µ-opioid system in a position to ameliorate aversive or defensive behavioral states associated with stress, withdrawal, physical pain or social rejection.
Collapse
|
227
|
Gandini MA, Souza IA, Raval D, Xu J, Pan YX, Zamponi GW. Differential regulation of Cav2.2 channel exon 37 variants by alternatively spliced μ-opioid receptors. Mol Brain 2019; 12:98. [PMID: 31775826 PMCID: PMC6880636 DOI: 10.1186/s13041-019-0524-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022] Open
Abstract
We have examined the regulation of mutually exclusive Cav2.2 exon 37a and b variants by the mouse μ-opioid receptor (mMOR) C-terminal splice variants 1, 1C and 1O in tsA-201 cells. Electrophysiological analyses revealed that both channel isoforms exhibit DAMGO-induced voltage-dependent (Gβγ-mediated) inhibition and its recovery by voltage pre-pulses, as well as a voltage-independent component. However, the two channel isoforms differ in their relative extent of voltage-dependent and independent inhibition, with Cav2.2-37b showing significantly more voltage-dependent inhibition upon activation of the three mMOR receptors studied. In addition, coexpression of either mMOR1 or mMOR1C results in an agonist-independent reduction in the peak current density of Cav2.2-37a channels, whereas the peak current density of Cav2.2-37b does not appear to be affected. Interestingly, this decrease is not due to an effect on channel expression at the plasma membrane, as demonstrated by biotinylation experiments. We further examined the mechanism underlying the agonist-independent modulation of Cav2.2-37a by mMOR1C. Incubation of cells with pertussis toxin did not affect the mMOR1C mediated inhibition of Cav2.2-37a currents, indicating a lack of involvement of Gi/o signaling. However, when a Src tyrosine kinase inhibitor was applied, the effect of mMOR1C was lost. Moreover, when we recorded currents using a Cav2.2-37a mutant in which tyrosine 1747 was replaced with phenylalanine (Y1747F), the agonist independent effects of mMOR1C were abolished. Altogether our findings show that Cav2.2-37a and Cav2.2-37b isoforms are subject to differential regulation by C-terminal splice variants of mMORs, and that constitutive mMOR1C activity and downstream tyrosine kinase activity exert a selective inhibition of the Cav2.2-37a splice variant, an N-type channel isoform that is highly enriched in nociceptors. Our study provides new insights into the roles of the MOR full-length C-terminal variants in modulating Cav2.2 channel isoform activities.
Collapse
Affiliation(s)
- Maria A Gandini
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ivana A Souza
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Dvij Raval
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jin Xu
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Ying-Xian Pan
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
228
|
Brutman JN, Sirohi S, Davis JF. Recent Advances in the Neurobiology of Altered Motivation Following Bariatric Surgery. Curr Psychiatry Rep 2019; 21:117. [PMID: 31707546 DOI: 10.1007/s11920-019-1084-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW There is compelling evidence in the clinical population that long-term weight loss secondary to bariatric surgery is mitigated by the reemergence of maladaptive feeding behaviors and in some cases new onset substance abuse. RECENT FINDINGS A review of the current literature suggests that physical restructuring of the GI tract during WLS alters secretion of feeding peptides and nutrient-sensing mechanisms that directly target the brain's endogenous reward system, the mesolimbic dopamine system. Post-surgical changes in GI physiology augment activation of the mesolimbic system. In some patients, this process may contribute to a reduced appetite for palatable food whereas in others it may support maladaptive motivated behavior for food and chemical drugs. It is concluded that future studies are required to detail the timing and duration of surgical-induced changes in GI-mesolimbic communication to more fully understand this phenomenon.
Collapse
Affiliation(s)
- Julianna N Brutman
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, 1815 Ferdinand's Lane, Pullman, WA, 99164, USA
| | - Sunil Sirohi
- Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA, USA
| | - Jon F Davis
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, 1815 Ferdinand's Lane, Pullman, WA, 99164, USA.
| |
Collapse
|
229
|
Le Rouzic V, Narayan A, Hunkle A, Marrone GF, Lu Z, Majumdar S, Xu J, Pan YX, Pasternak GW. Pharmacological Characterization of Levorphanol, a G-Protein Biased Opioid Analgesic. Anesth Analg 2019; 128:365-373. [PMID: 29649035 DOI: 10.1213/ane.0000000000003360] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Levorphanol is a potent analgesic that has been used for decades. Most commonly used for acute and cancer pain, it also is effective against neuropathic pain. The recent appreciation of the importance of functional bias and the uncovering of multiple µ opioid receptor splice variants may help explain the variability of patient responses to different opioid drugs. METHODS Here, we evaluate levorphanol in a variety of traditional in vitro receptor binding and functional assays. In vivo analgesia studies using the radiant heat tail flick assay explored the receptor selectivity of the responses through the use of knockout (KO) mice, selective antagonists, and viral rescue approaches. RESULTS Receptor binding studies revealed high levorphanol affinity for all the μ, δ, and κ opioid receptors. In S-GTPγS binding assays, it was a full agonist at most µ receptor subtypes, with the exception of MOR-1O, but displayed little activity in β-arrestin2 recruitment assays, indicating a preference for G-protein transduction mechanisms. A KO mouse and selective antagonists confirmed that levorphanol analgesia was mediated through classical µ receptors, but there was a contribution from 6 transmembrane targets, as illustrated by a lower response in an exon 11 KO mouse and its rescue with a virally transfected 6 transmembrane receptor splice variant. Compared to morphine, levorphanol had less respiratory depression at equianalgesic doses. CONCLUSIONS While levorphanol shares many of the same properties as the classic opioid morphine, it displays subtle differences that may prove helpful in its clinical use. Its G-protein signaling bias is consistent with its diminished respiratory depression, while its incomplete cross tolerance with morphine suggests it may prove valuable clinically with opioid rotation.
Collapse
Affiliation(s)
- Valerie Le Rouzic
- From the Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ankita Narayan
- From the Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amanda Hunkle
- From the Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gina F Marrone
- From the Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zhigang Lu
- First Clinical Medical College, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Susruta Majumdar
- From the Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jin Xu
- From the Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ying-Xian Pan
- From the Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gavril W Pasternak
- From the Department of Neurology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
230
|
Wu Z, Hruby VJ. Toward a Universal μ-Agonist Template for Template-Based Alignment Modeling of Opioid Ligands. ACS OMEGA 2019; 4:17457-17476. [PMID: 31656918 PMCID: PMC6812133 DOI: 10.1021/acsomega.9b02244] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/25/2019] [Indexed: 05/04/2023]
Abstract
Opioid ligands are a large group of G-protein-coupled receptor ligands possessing high structural diversity, along with complicated structure-activity relationships (SARs). To better understand their structural correlations as well as the related SARs, we developed the innovative template-based alignment modeling in our recent studies on a variety of opioid ligands. As previously reported, this approach showed promise but also with limitations, which was mainly attributed to the small size of morphine as a template. With this study, we set out to construct an artificial μ-agonist template to overcome this limitation. The newly constructed template contained a largely extended scaffold, along with a few special μ-features relevant to the μ-selectivity of opioid ligands. As demonstrated in this paper, the new template showed significantly improved efficacy in facilitating the alignment modeling of a wide variety of opioid ligands. This report comprises of two main parts. Part 1 discusses the general construction process and the structural features as well as a few typical examples of the template applications and Part 2 focuses on the template refinement and validation.
Collapse
Affiliation(s)
- Zhijun Wu
- ABC Resource, Plainsboro, New Jersey 08536, United States
- E-mail:
| | - Victor J. Hruby
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85716, United States
| |
Collapse
|
231
|
Murphy A, Barbaro J, Martínez-Aguado P, Chilunda V, Jaureguiberry-Bravo M, Berman JW. The Effects of Opioids on HIV Neuropathogenesis. Front Immunol 2019; 10:2445. [PMID: 31681322 PMCID: PMC6813247 DOI: 10.3389/fimmu.2019.02445] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022] Open
Abstract
HIV associated neurocognitive disorders (HAND) are a group of neurological deficits that affect approximately half of people living with HIV (PLWH) despite effective antiretroviral therapy (ART). There are currently no reliable molecular biomarkers or treatments for HAND. Given the national opioid epidemic, as well as illegal and prescription use of opioid drugs among PLWH, it is critical to characterize the molecular interactions between HIV and opioids in cells of the CNS. It is also important to study the role of opioid substitution therapies in the context of HIV and CNS damage in vitro and in vivo. A major mechanism contributing to HIV neuropathogenesis is chronic, low-level inflammation in the CNS. HIV enters the brain within 4–8 days after peripheral infection and establishes CNS reservoirs, even in the context of ART, that are difficult to identify and eliminate. Infected cells, including monocytes, macrophages, and microglia, produce chemokines, cytokines, neurotoxic mediators, and viral proteins that contribute to chronic inflammation and ongoing neuronal damage. Opioids have been shown to impact these immune cells through a variety of molecular mechanisms, including opioid receptor binding and cross desensitization with chemokine receptors. The effects of opioid use on cognitive outcomes in individuals with HAND in clinical studies is variable, and thus multiple biological mechanisms are likely to contribute to the complex relationship between opioids and HIV in the CNS. In this review, we will examine what is known about both HIV and opioid mediated neuropathogenesis, and discuss key molecular processes that may be impacted by HIV and opioids in the context of neuroinflammation and CNS damage. We will also assess what is known about the effects of ART on these processes, and highlight areas of study that should be addressed in the context of ART.
Collapse
Affiliation(s)
- Aniella Murphy
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - John Barbaro
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Pablo Martínez-Aguado
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Vanessa Chilunda
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Matias Jaureguiberry-Bravo
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Joan W Berman
- Laboratory of Dr. Joan W. Berman, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States.,Laboratory of Dr. Joan W. Berman, Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
232
|
Porter MR, Xiao H, Wang J, Smith SB, Topczewski JJ. 3-Amino-chromanes and Tetrahydroquinolines as Selective 5-HT 2B, 5-HT 7, or σ 1 Receptor Ligands. ACS Med Chem Lett 2019; 10:1436-1442. [PMID: 31620230 DOI: 10.1021/acsmedchemlett.9b00225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/23/2019] [Indexed: 12/26/2022] Open
Abstract
The phenethylamine backbone is a privileged substructure found in a wide variety of G protein-coupled receptor (GPCR) ligands. This includes both endogenous neurotransmitters and active pharmaceutical agents. More than 20 structurally unique heterocyclic phenethylamine derivatives were broadly evaluated for GPCR affinity. Selective ligands for the 5-HT2B, 5-HT7, and σ1 receptors were identified, each with low nanomolar binding affinities. The σ1 receptor affinity was supported in a cellular assay that provided evidence for increased cell survival under oxidative stress.
Collapse
Affiliation(s)
- Matthew R. Porter
- Department of Chemistry, University of Minnesota Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia 30912, United States
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia 30912, United States
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia 30912, United States
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, United States
| | - Joseph J. Topczewski
- Department of Chemistry, University of Minnesota Twin Cities, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
233
|
Bellamy JR, Rubin BR, Zverovich A, Zhou Y, Contoreggi NH, Gray JD, McEwen BS, Kreek MJ, Milner TA. Sex and chronic stress differentially alter phosphorylated mu and delta opioid receptor levels in the rat hippocampus following oxycodone conditioned place preference. Neurosci Lett 2019; 713:134514. [PMID: 31560995 DOI: 10.1016/j.neulet.2019.134514] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 09/21/2019] [Indexed: 01/16/2023]
Abstract
Following oxycodone conditioned place preference (CPP) in naïve female and male Sprague Dawley rats, delta- and mu-opioid receptors (DORs and MORs) redistribute in hippocampal CA3 pyramidal cells and GABAergic interneurons in a manner that would promote opioid-associative learning processes, particularly in females. MORs and DORs similarly redistribute in CA3 and hilar neurons following chronic immobilization stress (CIS) in females, but not males, essentially "priming" the opioid system for oxycodone-associative learning. Following CIS, only females acquire oxycodone CPP. The present study determined whether sex and CIS differentially affect the levels of phosphorylated MORs and DORs (pMORs and pDORs) in the hippocampus following oxycodone CPP as phosphorylation is important for opioid receptor internationalization and trafficking. In naïve oxycodone-injected (Oxy) female rats, the density of pMOR-immunoreactivity (ir) was increased in CA1 stratum oriens and CA3a,b strata lucidum and radiatum compared to saline-injected (Sal)-females. Additionally, the density of pDOR-ir increased in the pyramidal cell layer and stratum radiatum of CA2/3a in Oxy-males compared to Sal-males. In CIS females that acquire CPP, pDOR-ir levels were increased in the CA2/3a. These findings indicate only rats that acquire oxycodone CPP have activated MORs and DORs in the hippocampus but that the subregion containing activated opioid receptors differs in females and males. These results are consistent with previously observed sex differences in the hippocampal opioid system following Oxy-CPP.
Collapse
Affiliation(s)
- Julia R Bellamy
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Batsheva R Rubin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Angelica Zverovich
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Natalina H Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States
| | - Jason D Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, United States; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, United States.
| |
Collapse
|
234
|
Rosas R, Huang XP, Roth BL, Dockendorff C. β-Fluorofentanyls Are pH-Sensitive Mu Opioid Receptor Agonists. ACS Med Chem Lett 2019; 10:1353-1356. [PMID: 31531209 DOI: 10.1021/acsmedchemlett.9b00335] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 08/13/2019] [Indexed: 02/08/2023] Open
Abstract
The concept recently postulated by Stein and co-workers (Science 2017, 355, 966) that mu opioid receptor (MOR) agonists possessing amines with attenuated basicity show pH-dependent activity and can selectively act at damaged, low pH tissues has been additionally supported by in vitro studies reported here. We synthesized and tested analogs of fentanyl possessing one or two fluorine atoms at the beta position of the phenethylamine side chain, with additional fluorines optionally added to the benzene ring of the side chain. These compounds were synthesized in 1 to 3 steps from commercial building blocks. The novel bis-fluorinated analog RR-49 showed superior pH sensitivity, with full efficacy relative to DAMGO, but with 19-fold higher potency (IC50) in a MOR cAMP assay at pH 6.5 versus 7.4. Such compounds hold significant promise as analgesics for inflammatory pain with reduced abuse potential.
Collapse
Affiliation(s)
- Ricardo Rosas
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, Wisconsin 53201-1881, United States
| | - Xi-Ping Huang
- National Institute of Mental Health Psychoactive Drug Screening Program, Department of Pharmacology, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599-7365, United States
| | - Bryan L. Roth
- National Institute of Mental Health Psychoactive Drug Screening Program, Department of Pharmacology, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599-7365, United States
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599-7365, United States
| | - Chris Dockendorff
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, Wisconsin 53201-1881, United States
| |
Collapse
|
235
|
Zou Y, Ewalt J, Ng HL. Recent Insights from Molecular Dynamics Simulations for G Protein-Coupled Receptor Drug Discovery. Int J Mol Sci 2019; 20:E4237. [PMID: 31470676 PMCID: PMC6747122 DOI: 10.3390/ijms20174237] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are critical drug targets. GPCRs convey signals from the extracellular to the intracellular environment through G proteins. Some ligands that bind to GPCRs activate different downstream signaling pathways. G protein activation, or -arrestin biased signaling, involves ligands binding to receptors and stabilizing conformations that trigger a specific pathway. -arrestin biased signaling has become a hot target for structure-based drug discovery. However, challenges include that there are few crystal structures available in the Protein Data Bank and that GPCRs are highly dynamic. Hence, molecular dynamics (MD) simulations are especially valuable for obtaining detailed mechanistic information, including identification of allosteric sites and understanding modulators' interactions with receptors and ligands. Here, we highlight recent MD simulation studies and enhanced sampling methods used to study biased G protein-coupled receptor signaling and their conformational dynamics as well as applications to drug discovery.
Collapse
Affiliation(s)
- Ye Zou
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - John Ewalt
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Ho-Leung Ng
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
236
|
Liu XY, Ginosar Y, Yazdi J, Hincker A, Chen ZF. Cross-talk between Human Spinal Cord μ-opioid Receptor 1Y Isoform and Gastrin-releasing Peptide Receptor Mediates Opioid-induced Scratching Behavior. Anesthesiology 2019; 131:381-391. [PMID: 31314749 PMCID: PMC7098053 DOI: 10.1097/aln.0000000000002776] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Although spinal opioids are safe and effective, pruritus is common and distressing. The authors previously demonstrated in mouse spinal cord that interactions between μ-opioid receptor isoform 1D and gastrin releasing peptide receptor mediate morphine-induced scratch. The C-terminal of 1D inhibits morphine-induced scratch without affecting analgesia. The authors hypothesize that human spinal cord also contains itch-specific μ-opioid receptor isoforms which interact with gastrin releasing peptide receptor. METHODS Reverse transcription polymerase chain reaction was performed on human spinal cord complimentary DNA from two human cadavers. Calcium responses to morphine (1 μM) were examined using calcium imaging microscopy on human cells (HEK293) coexpressing gastrin releasing peptide receptor and different human μ-opioid receptor isoforms. The authors assessed morphine-induced scratching behavior and thermal analgesia in mice following intrathecal injection of morphine (0.3 nmol) and a transactivator of transcription peptide designed from C-terminal sequences of 1Y isoform (0, 0.1, and 0.4 nmol). RESULTS The authors demonstrated 1Y expression in the spinal cord dorsal horn. Morphine administration evoked a calcium response (mean ± SD) (57 ± 13 nM) in cells coexpressing both gastrin releasing peptide receptor and the 1Y isomer. This was blocked by 10 μM naltrexone (0.7 ± 0.4 nM; P < 0.0001), 1 μM gastrin-releasing peptide receptor antagonist (3 ± 2 nM; P < 0.0001), or 200 μM 1Y-peptide (2 + 2 nM; P < 0.0001). In mice, 0.4 nmol 1Y-peptide significantly attenuated morphine-induced scratching behaviors (scratching bouts, vehicle vs. 1Y-peptide) (92 ± 31 vs. 38 ± 29; P = 0.011; n = 6 to 7 mice per group), without affecting morphine antinociception in warm water tail immersion test (% of maximum possible effect) (70 ± 21 vs. 67 ± 22; P = 0.80; n = 6 mice per group). CONCLUSIONS Human μ-opioid receptor 1Y isomer is a C-terminal splicing variant of Oprm1 gene identified in human spinal cord. Cross-talk between 1Y and gastrin releasing peptide receptor is required for mediating opioid-induced pruritus. Disrupting the cross talk may have implications for therapeutic uncoupling of desired analgesic effects from side effects of opioids.
Collapse
Affiliation(s)
- Xian-Yu Liu
- From the Center for the Study of Itch, Departments of Anesthesiology, Psychiatry and Developmental Biology (X.-Y.L., Z.-F.C.) the Division of Obstetric Anesthesiology, Department of Anesthesiology, Barnes Jewish Hospital (Y.G., A.H.), Washington University School of Medicine, St. Louis, Missouri the Mother and Child Anesthesia Unit, Department of Anesthesiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel (Y.G.) SpineMore Surgical Associates, St. Louis, Missouri (J.Y.)
| | | | | | | | | |
Collapse
|
237
|
Irie T, Shum R, Deni I, Hunkele A, Le Rouzic V, Xu J, Wilson R, Fischer GW, Pasternak GW, Pan YX. Identification of Abundant and Evolutionarily Conserved Opioid Receptor Circular RNAs in the Nervous System Modulated by Morphine. Mol Pharmacol 2019; 96:247-258. [PMID: 31243060 DOI: 10.1124/mol.118.113977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 06/03/2019] [Indexed: 01/25/2023] Open
Abstract
Circular RNAs (circRNAs) are a distinct category of single-stranded, covalently closed RNAs formed by backsplicing. The functions of circRNAs are incompletely known and are under active investigation. Here, we report that in addition to traditional linear mRNAs (linRNA), mouse, rat, and human opioid receptor genes generate exonic circRNA isoforms. Using standard molecular biologic methods, Oprm1 circRNAs (circOprm1) were detected in RNAs of rodent and human brains and spinal cords, as well as human neuroblastoma cells, suggesting evolutionary conservation. Sequencing confirmed backsplicing using canonical splice sites. Oprm1 circRNAs were sense-stranded circRNAs resistant to RNase R digestion. The relative abundance of Oprm1 circRNA to linRNA determined by quantitative reverse transcription polymerase chain reaction varied among mouse brain regions, with circRNA isoforms predominating in rostral structures and less abundant in brain stem. Chronic morphine exposure in mice increased brain circOprm1e2.3 and circOprm1.e2.e3.e4(302) levels by 1.5- to 1.6-fold relative to linRNA. Sequence analysis predicted numerous microRNA binding sites within Oprm1 circRNA sequences, suggesting a potential role in microRNA sequestration through sponging. In addition, we observed that other opioid receptor genes including δ, κ, and nociceptin receptor genes produced similar circRNAs. In conclusion, all members of the opioid receptor gene family express circRNAs, with Oprm1 circRNA levels exceeding those of linear forms in some regions. SIGNIFICANCE STATEMENT: The modulation of Oprm1 circular RNA (circRNA) expression by morphine, coupled with the high abundance and existence of potential miRNA binding sites with circRNA sequences suggests the potential role of Oprm1 circRNAs in chronic opioid effects such as tolerance.
Collapse
Affiliation(s)
- Takeshi Irie
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Rebecca Shum
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ioanna Deni
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Amanda Hunkele
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Valerie Le Rouzic
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jin Xu
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Roger Wilson
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Gregory W Fischer
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Gavril W Pasternak
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ying-Xian Pan
- Departments of Anesthesiology and Critical Care Medicine (T.I., R.W., G.W.F.) and Neurology (G.W.P., Y.-X.P.), and Molecular Pharmacology Program (R.S., I.D., A.H., V.L.R., J.X., G.W.P.), Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
238
|
Nielsen S, Crossin R, Middleton M, Martin C, Wilson J, Lam T, Scott D, Smith K, Lubman D. Comparing rates and characteristics of ambulance attendances related to extramedical use of pharmaceutical opioids in Australia: a protocol for a retrospective observational study. BMJ Open 2019; 9:e029170. [PMID: 31138584 PMCID: PMC6549600 DOI: 10.1136/bmjopen-2019-029170] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/11/2019] [Accepted: 04/17/2019] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION AND AIMS Extramedical use of, and associated harms with pharmaceutical opioids are common. Analysis of coded ambulance clinical records provides a unique opportunity to examine a national population-level indicator of relative harms. This protocol describes an observational study with three aims: (1) to compare supply adjusted rates of pharmaceutical opioid-related ambulance attendances for buprenorphine, codeine, fentanyl, oxycodone, oxycodone-naloxone, morphine, pethidine, tramadol and tapentadol; (2) to compare presentation characteristics for these commonly used pharmaceutical opioids and (3) to describe the context surrounding ambulance presentations related to oxycodone, a widely used opioid with an established abuse liability, and tapentadol, a more recent 'atypical' opioid on the Australian market, with fewer studies that have directly examined signals of extramedical use. METHOD Trained coders extract data from clinical records for ambulance presentations relating to extramedical use of commonly used pharmaceutical opioids. These data form the basis of a large, national database that captures alcohol-related and drug-related harms. Supply adjusted rates of presentations will be examined using Poisson regression. Multinomial logistic regression will be used to compare severity and other characteristics of attendances relating to different pharmaceutical opioids. Tapentadol-related and oxycodone-related cases will be qualitatively examined to understand the situationally specific contexts of the ambulance attendances outside of the characteristics captured in routinely coded variables. ETHICS AND DISSEMINATION Ethics approval related to analysis of ambulance attendance data was obtained from the Eastern Health Human Research Ethics Committee (E122 08-09), with an amendment specific to the qualitative analysis. Findings will be submitted for peer review in 2019. The understanding of risk profiles in real-world settings is of international public health importance. The analysis and publication of findings from this national dataset of clinical records will provide one of the most nuanced analyses to date of relative harms across nine pharmaceutical opioids over a 6-year period.
Collapse
Affiliation(s)
- Suzanne Nielsen
- Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
- National Drug and Alcohol Research Centre, UNSW Sydney, Randwick, New South Wales, Australia
- Turning Point, Monash University Eastern Health Clinical School, North Richmond, Victoria, Australia
| | - Rose Crossin
- Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
- Turning Point, Monash University Eastern Health Clinical School, North Richmond, Victoria, Australia
| | - Melissa Middleton
- Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Catherine Martin
- Biostatistics Unit, Public Health and Preventative Medicine, Monash University, Melbourne, New South Wales, Australia
| | - James Wilson
- Turning Point, Monash University Eastern Health Clinical School, North Richmond, Victoria, Australia
| | - Tina Lam
- Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Debbie Scott
- Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
- Turning Point, Monash University Eastern Health Clinical School, North Richmond, Victoria, Australia
| | - Karen Smith
- Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
- Centre for Research and Evaluation, Ambulance Victoria, Doncaster, Victoria, Australia
- Department of Community Emergency Health and Paramedic Practice, Monash University, Frankston, Victoria, Australia
| | - Dan Lubman
- Monash Addiction Research Centre, Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
- Turning Point, Monash University Eastern Health Clinical School, North Richmond, Victoria, Australia
| |
Collapse
|
239
|
Molecular dynamics of fentanyl bound to μ-opioid receptor. J Mol Model 2019; 25:144. [DOI: 10.1007/s00894-019-3999-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 03/21/2019] [Indexed: 12/17/2022]
|
240
|
Calvo F, Almada RC, Dos Anjos-Garcia T, Falconi-Sobrinho LL, Paschoalin-Maurin T, Bazaglia-de-Sousa G, Medeiros P, Silva JAD, Lobão-Soares B, Coimbra NC. Panicolytic-like effect of µ 1-opioid receptor blockade in the inferior colliculus of prey threatened by Crotalus durissus terrificus pit vipers. J Psychopharmacol 2019; 33:577-588. [PMID: 30663473 DOI: 10.1177/0269881118822078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The endogenous opioid peptide system has been implicated in the neural modulation of fear and anxiety organised by the dorsal midbrain. Furthermore, previous results indicate a fundamental role played by inferior colliculus (IC) opioid mechanisms during the expression of defensive behaviours, but the involvement of the IC µ1-opioid receptor in the modulation of anxiety- and panic attack-related behaviours remains unclear. Using a prey-versus-snake confrontation paradigm, we sought to investigate the effects of µ1-opioid receptor blockade in the IC on the defensive behaviour displayed by rats in a dangerous situation. METHODS Specific pathogen-free Wistar rats were treated with microinjection of the selective µ1-opioid receptor antagonist naloxonazine into the IC at different concentrations (1.0, 3.0 and 5.0 µg/0.2 µL) and then confronted with rattlesnakes ( Crotalus durissus terrificus). The defensive behavioural repertoire, such as defensive attention, flat back approach (FBA), startle, defensive immobility, escape or active avoidance, displayed by rats either during the confrontations with wild snakes or during re-exposure to the experimental context without the predator was analysed. RESULTS The blockade of µ1-opioid receptors in the IC decreased the expression of both anxiety-related behaviours (defensive attention, FBA) and panic attack-related responses (startle, defensive immobility and escape) during the confrontation with rattlesnakes. A significant decrease in defensive attention was also recorded during re-exposure of the prey to the experimental apparatus context without the predator. CONCLUSION Taken together, these results suggest that a decrease in µ1-opioid receptor signalling activity within the IC modulates anxiety- and panic attack-related behaviours in dangerous environments.
Collapse
Affiliation(s)
- Fabrício Calvo
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,2 Department of Pharmacology, São Lucas College, Porto Velho (RO), Brazil.,3 Aparício Carvalho Integrative College (FIMCA), Porto Velho (RO), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil
| | - Rafael Carvalho Almada
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,5 Behavioural Neurosciences Institute (INeC), Ribeirão Preto (SP), Brazil
| | - Tayllon Dos Anjos-Garcia
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,6 NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil
| | - Luiz Luciano Falconi-Sobrinho
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,5 Behavioural Neurosciences Institute (INeC), Ribeirão Preto (SP), Brazil.,6 NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil
| | - Tatiana Paschoalin-Maurin
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,5 Behavioural Neurosciences Institute (INeC), Ribeirão Preto (SP), Brazil
| | - Guilherme Bazaglia-de-Sousa
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,5 Behavioural Neurosciences Institute (INeC), Ribeirão Preto (SP), Brazil.,6 NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil
| | - Priscila Medeiros
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,5 Behavioural Neurosciences Institute (INeC), Ribeirão Preto (SP), Brazil
| | - Juliana Almeida da Silva
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,5 Behavioural Neurosciences Institute (INeC), Ribeirão Preto (SP), Brazil.,6 NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil
| | - Bruno Lobão-Soares
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,5 Behavioural Neurosciences Institute (INeC), Ribeirão Preto (SP), Brazil.,7 Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte (UFRN), Natal (RN), Brazil
| | - Norberto Cysne Coimbra
- 1 Department of Pharmacology, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,4 Ophidiarium LNN-FMRP-USP/INeC, University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brasil.,5 Behavioural Neurosciences Institute (INeC), Ribeirão Preto (SP), Brazil.,6 NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil
| |
Collapse
|
241
|
Eidson LN, Murphy AZ. Inflammatory mediators of opioid tolerance: Implications for dependency and addiction. Peptides 2019; 115:51-58. [PMID: 30890355 PMCID: PMC6863079 DOI: 10.1016/j.peptides.2019.01.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 12/27/2022]
Abstract
Each year, over 50 million Americans suffer from persistent pain, including debilitating headaches, joint pain, and severe back pain. Although morphine is amongst the most effective analgesics available for the management of severe pain, prolonged morphine treatment results in decreased analgesic efficacy (i.e., tolerance). Despite significant headway in the field, the mechanisms underlying the development of morphine tolerance are not well understood. The midbrain ventrolateral periaqueductal gray (vlPAG) is a primary neural substrate for the analgesic effects of morphine, as well as for the development of morphine tolerance. A growing body of literature indicates that activated glia (i.e., microglia and astrocytes) facilitate pain transmission and oppose morphine analgesia, making these cells important potential targets in the treatment of chronic pain. Morphine affects glia by binding to the innate immune receptor toll-like receptor 4 (TLR4), leading to the release of proinflammatory cytokines and opposition of morphine analgesia. Despite the established role of the vlPAG as an integral locus for the development of morphine tolerance, most studies have examined the role of glia activation within the spinal cord. Additionally, the role of TLR4 in the development of tolerance has not been elucidated. This review attempts to summarize what is known regarding the role of vlPAG glia and TLR4 in the development of morphine tolerance. These data, together, provide information about the mechanism by which central nervous system glia regulate morphine tolerance, and identify a potential therapeutic target for the enhancement of analgesic efficacy in the clinical treatment of chronic pain.
Collapse
Affiliation(s)
- Lori N Eidson
- Department of Physiology, Emory University, Atlanta, GA, 30322, United States
| | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30308, United States.
| |
Collapse
|
242
|
Vicario N, Pasquinucci L, Spitale FM, Chiechio S, Turnaturi R, Caraci F, Tibullo D, Avola R, Gulino R, Parenti R, Parenti C. Simultaneous Activation of Mu and Delta Opioid Receptors Reduces Allodynia and Astrocytic Connexin 43 in an Animal Model of Neuropathic Pain. Mol Neurobiol 2019; 56:7338-7354. [PMID: 31030416 DOI: 10.1007/s12035-019-1607-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/12/2019] [Indexed: 12/27/2022]
Abstract
Neuropathic pain is a chronic condition triggered by lesions to the somatosensory nervous system in which pain stimuli occur spontaneously or as pathologically amplified responses. In this scenario, the exchange of signaling molecules throughout cell-to-cell and cell-to-extracellular environment communications plays a key role in the transition from acute to chronic pain. As such, connexin 43 (Cx43), the core glial gap junction and hemichannel-forming protein, is considered a triggering factor for disease chronicization in the central nervous system (CNS). Drugs targeting μ opioid receptors (MOR) are currently used for moderate to severe pain conditions, but their use in chronic pain is limited by the tolerability profile. δ opioid receptors (DOR) have become attractive targets for the treatment of persistent pain and have been associated with the inhibition of pain-sustaining factors. Moreover, it has been shown that simultaneous targeting of MOR and DOR leads to an improved pharmacological fingerprint. Herein, we aimed to study the effects of the benzomorphan ligand LP2, a multitarget MOR/DOR agonist, in an experimental model of neuropathic pain induced by the unilateral sciatic nerve chronic constriction injury (CCI) on male Sprague-Dawley rats. Results showed that LP2 significantly ameliorated mechanical allodynia from the early phase of treatment up to 21 days post-ligatures. We additionally showed that LP2 prevented CCI-induced Cx43 alterations and pro-apoptotic signaling in the CNS. These findings increase the knowledge of neuropathic pain development and the role of spinal astrocytic Cx43, suggesting new approaches for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123, Catania, Italy
| | - Lorella Pasquinucci
- Department of Drug Sciences, Section of Medicinal Chemistry, University of Catania, 95125, Catania, Italy
| | - Federica M Spitale
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123, Catania, Italy
| | - Santina Chiechio
- Department of Drug Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125, Catania, Italy.,Oasi Research Institute-IRCCS, 94018, Troina, Italy
| | - Rita Turnaturi
- Department of Drug Sciences, Section of Medicinal Chemistry, University of Catania, 95125, Catania, Italy
| | - Filippo Caraci
- Department of Drug Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125, Catania, Italy.,Oasi Research Institute-IRCCS, 94018, Troina, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123, Catania, Italy
| | - Roberto Avola
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123, Catania, Italy
| | - Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95123, Catania, Italy.
| | - Carmela Parenti
- Department of Drug Sciences, Section of Pharmacology and Toxicology, University of Catania, 95125, Catania, Italy
| |
Collapse
|
243
|
Calhoun SE, Meunier CJ, Lee CA, McCarty GS, Sombers LA. Characterization of a Multiple-Scan-Rate Voltammetric Waveform for Real-Time Detection of Met-Enkephalin. ACS Chem Neurosci 2019; 10:2022-2032. [PMID: 30571911 PMCID: PMC6473485 DOI: 10.1021/acschemneuro.8b00351] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023] Open
Abstract
Opioid peptides are critically involved in a variety of physiological functions necessary for adaptation and survival, and as such, understanding the precise actions of endogenous opioid peptides will aid in identification of potential therapeutic strategies to treat a variety of disorders. However, few analytical tools are currently available that offer both the sensitivity and spatial resolution required to monitor peptidergic concentration fluctuations in situ on a time scale commensurate with that of neuronal communication. Our group has developed a multiple-scan-rate waveform to enable real-time voltammetric detection of tyrosine containing neuropeptides. Herein, we have evaluated the waveform parameters to increase sensitivity to methionine-enkephalin (M-ENK), an endogenous opioid neuropeptide implicated in pain, stress, and reward circuits. M-ENK dynamics were monitored in adrenal gland tissue, as well as in the dorsal striatum of anesthetized and freely behaving animals. The data reveal cofluctuations of catecholamine and M-ENK in both locations and provide measurements of M-ENK dynamics in the brain with subsecond temporal resolution. Importantly, this work also demonstrates how voltammetric waveforms can be customized to enhance detection of specific target analytes, broadly speaking.
Collapse
Affiliation(s)
- S. E. Calhoun
- Department of Chemistry, North
Carolina State University, Raleigh, North Carolina 27695, United States
| | - C. J. Meunier
- Department of Chemistry, North
Carolina State University, Raleigh, North Carolina 27695, United States
| | - C. A. Lee
- Department of Chemistry, North
Carolina State University, Raleigh, North Carolina 27695, United States
| | - G. S. McCarty
- Department of Chemistry, North
Carolina State University, Raleigh, North Carolina 27695, United States
| | - L. A. Sombers
- Department of Chemistry, North
Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
244
|
Moyano J, Aguirre L. Opioids in the immune system: from experimental studies to clinical practice. ACTA ACUST UNITED AC 2019; 65:262-269. [PMID: 30892453 DOI: 10.1590/1806-9282.65.2.262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/27/2018] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Opioids interact with both innate and adaptive immune systems and have direct effects on opioid receptors located on immune cells. Research on this topic has provided evidence of the opioid influence on the immune response associated with surgical stress. The immunological effects of opioids are currently being investigated, particularly whether they influence the outcome of surgery or the underlying disease regarding important aspects like infection or cancer progression. This review addresses background research related to the influence of the opioid receptor on the immune system, the immunosuppressive effect associated with major opioids during the perioperative period, and their clinical relevance. The objective of the study was to review the effects of opioids on the immune system.
Collapse
Affiliation(s)
- Jairo Moyano
- Anesthesia Department, Pain Service, Hospital Universitario Fundación Santafé de Bogotá, Bogotá, Colombia
| | - Luisa Aguirre
- Anesthesia Department, Pain Service, Hospital Universitario Fundación Santafé de Bogotá, Bogotá, Colombia
| |
Collapse
|
245
|
Vassoler FM, Toorie AM, Byrnes EM. Increased cocaine reward in offspring of females exposed to morphine during adolescence. Psychopharmacology (Berl) 2019; 236:1261-1272. [PMID: 30506236 DOI: 10.1007/s00213-018-5132-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
Abstract
RATIONALE A growing body of evidence demonstrates that environmental exposures can impact the physiology and behavior of subsequent generations. We have previously demonstrated reduced morphine self-administration in the F1 and F2 offspring of female rats exposed to morphine during adolescence. OBJECTIVES The current study was designed to determine whether attenuated self-administration for a substance not in the opioid class is also observed in the F1 progeny of adolescent morphine exposed females. METHODS Female adolescent rats were administered morphine at increasing doses for 10 days (P30-39). Females then remained drug free for at least 3 weeks prior to mating with drug-naïve males. As adults, male and female offspring (F1 animals) were tested for cocaine self-administration acquisition, progressive ratio, extinction, and reinstatement. In addition, β-endorphin peptide levels were measured in the nucleus accumbens (NAc) of behaviorally experienced animals following reinstatement and in behaviorally naïve littermates after acute cocaine (0 or 10 mg/kg, i.p.). Proopiomelanocortin, the polypeptide that is cleaved to produce β-endorphin, as well as β-endorphin, was examined in the arcuate nucleus of the hypothalamus and the nucleus accumbens, respectively. Finally, corticosterone was measured following acute cocaine. RESULTS While no differences were observed during the cocaine acquisition phase (FR-1 and FR-5 schedules), under a PR schedule, Mor-F1 animals (both males and females) had increased motivated responding for cocaine. In addition, Mor-F1 males demonstrated enhanced reinstatement compared to Sal-F1 males. In Mor-F1 males, an acute injection of cocaine (10 mg/kg, i.p.) decreased β-endorphin levels in the NAc compared to a saline injection while acute cocaine increased β-endorphin in the NAc in Sal-F1 males compared to saline injection. Following acute cocaine, Mor-F1 males had significantly lower levels of β-endorphin in the Nac compared to Sal-F1 males. Additionally, β-endorphin levels in the nucleus accumbens were negatively correlated with reinstatement behavior only in Mor-F1 males. Levels of POMC in the arcuate nucleus were elevated in Mor-F1 males compared to Sal-F1 males, a main effect driven primarily by POMC levels in the acute cocaine condition. These changes were not observed in Mor-F1 females. Finally, plasma corticosterone was increased in Mor-F1 males regardless of acute injection while Mor-F1 females displayed increased corticosterone in response to acute cocaine. CONCLUSIONS These data indicate that morphine prior to conception increases the rewarding effects of cocaine in male and female offspring. In addition, sex-specific alterations in endogenous opioids and hypothalamic physiology were observed.
Collapse
Affiliation(s)
- Fair M Vassoler
- Tufts University Cummings School of Veterinary Medicine, 200 Westboro Road, North Grafton, MA, 01536, USA.
| | - Anika M Toorie
- Tufts University Cummings School of Veterinary Medicine, 200 Westboro Road, North Grafton, MA, 01536, USA
| | - Elizabeth M Byrnes
- Tufts University Cummings School of Veterinary Medicine, 200 Westboro Road, North Grafton, MA, 01536, USA
| |
Collapse
|
246
|
Abstract
Pain has a useful protective role; through avoidance learning, it helps to decrease the probability of engaging in tissue-damaging, or otherwise dangerous experiences. In our modern society, the experience of acute post-surgical pain and the development of chronic pain states represent an unnecessary negative outcome. This has become an important health issue as more than 30% of the US population reports experiencing "unnecessary" pain at any given time. Opioid therapies are often efficacious treatments for severe and acute pain; however, in addition to their powerful analgesic properties, opioids produce potent reinforcing properties and their inappropriate use has led to the current opioid overdose epidemic in North America. Dissecting the allostatic changes occurring in nociceptors and neuronal pathways in response to pain are the first and most important steps in understanding the physiologic changes underlying the opioid epidemic. Full characterization of these adaptations will provide novel targets for the development of safer pharmacotherapies. In this review, we highlight the current efforts toward safer opioid treatments and describe our current knowledge of the interaction between pain and opioid systems.
Collapse
Affiliation(s)
- Nicolas Massaly
- Department of Anesthesiology; Washington University in St. Louis; St. Louis, MO, 63110 ; USA
- Washington University Pain Center; St. Louis, MO, 63110 ; USA
- Washington University in St Louis; School of Medicine; St. Louis, MO, 63110 ; USA
| | - Jose A Morón
- Department of Anesthesiology; Washington University in St. Louis; St. Louis, MO, 63110 ; USA
- Washington University Pain Center; St. Louis, MO, 63110 ; USA
- Washington University in St Louis; School of Medicine; St. Louis, MO, 63110 ; USA
- Department of Neuroscience; Washington University in St. Louis; St. Louis, MO, 63110 ; USA
| |
Collapse
|
247
|
Obeng S, Wang H, Jali A, Stevens DL, Akbarali HI, Dewey WL, Selley DE, Zhang Y. Structure-Activity Relationship Studies of 6α- and 6β-Indolylacetamidonaltrexamine Derivatives as Bitopic Mu Opioid Receptor Modulators and Elaboration of the "Message-Address Concept" To Comprehend Their Functional Conversion. ACS Chem Neurosci 2019; 10:1075-1090. [PMID: 30156823 PMCID: PMC6405326 DOI: 10.1021/acschemneuro.8b00349] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Structure-activity relationship (SAR) studies of numerous opioid ligands have shown that introduction of a methyl or ethyl group on the tertiary amino group at position 17 of the epoxymorphinan skeleton generally results in a mu opioid receptor (MOR) agonist while introduction of a cyclopropylmethyl group typically leads to an antagonist. Furthermore, it has been shown that introduction of heterocyclic ring systems at position 6 can favor antagonism. However, it was reported that 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-[(2'-indolyl)acetamido]morphinan (INTA), which bears a cyclopropylmethyl group at position 17 and an indole ring at position 6, acted as a MOR agonist. We herein report a SAR study on INTA with a series of its complementary derivatives to understand how introduction of an indole moiety with α or β linkage at position 6 of the epoxymorphinan skeleton may influence ligand function. Interestingly, one of INTA derivatives, compound 15 (NAN) was identified as a MOR antagonist both in vitro and in vivo. Molecular modeling studies revealed that INTA and NAN may interact with different domains of the MOR allosteric binding site. In addition, INTA may interact with W293 and N150 residues found in the orthosteric site to stabilize MOR activation conformation while NAN does not. These results suggest that INTA and NAN may be bitopic ligands and the type of allosteric interactions with the MOR influence their functional activity. These insights along with our enriched comprehension of the "message-address" concept will to benefit future ligand design.
Collapse
MESH Headings
- Allosteric Regulation/drug effects
- Allosteric Regulation/physiology
- Analgesics, Opioid/chemistry
- Analgesics, Opioid/pharmacology
- Animals
- CHO Cells
- Cricetinae
- Cricetulus
- Dose-Response Relationship, Drug
- Male
- Mice
- Narcotic Antagonists/chemistry
- Narcotic Antagonists/pharmacology
- Protein Binding/drug effects
- Protein Binding/physiology
- Protein Structure, Secondary
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/physiology
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Samuel Obeng
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Huiqun Wang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Abdulmajeed Jali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - David L. Stevens
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Hamid I. Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - William L. Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Dana E. Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| |
Collapse
|
248
|
Mercado-Reyes J, Almanza A, Segura-Chama P, Pellicer F, Mercado F. D2-like receptor agonist synergizes the μ-opioid agonist spinal antinociception in nociceptive, inflammatory and neuropathic models of pain in the rat. Eur J Pharmacol 2019; 853:56-64. [PMID: 30876975 DOI: 10.1016/j.ejphar.2019.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/25/2019] [Accepted: 03/12/2019] [Indexed: 11/19/2022]
Abstract
Opioids are potent analgesic drugs, but their use has been limited due to their side effects. Antinociceptive effects of D2-like receptor agonists such as quinpirole have been shown at the spinal cord level; however, their efficacy is not as high as that of opioids. Dopaminergic agonists are long-prescribed and well-tolerated drugs that have been useful to treat clinically and experimentally painful conditions. Because current pain treatments are not completely effective, the aim of this work was to determine if a D2-like receptor agonist improves the antinociceptive effects of a μ-opioid receptor agonist. Drugs were intrathecally administered in adult rats; mechanonociceptive and thermonociceptive tests were carried out. Intraplantar injection of complete Freund's adjuvant (CFA) and sciatic loose ligation (SLL) were used for inflammatory and neuropathic models of pain, respectively. In intact animals, D-Ala2, N-MePhe4, Gly-ol-enkephalin (DAMGO; a µ-opioid receptor agonist) increased the paw withdrawal latencies (PWL) in thermal and mechanical nociceptive tests in a dose-dependent manner. Quinpirole (D2-like receptor agonist) increased PWL only in mechanonociception. In the presence of quinpirole (1 nmol), the ED50 of the mechanical antinociceptive effect of DAMGO was significantly decreased (8-fold). Coadministration of 1 nmol quinpirole and 30 pmol DAMGO completely reversed hyperalgesia in the CFA model, whereas 100 pmol DAMGO plus 1 nmol quinpirole reversed the allodynia in the SLL model. This work offers evidence about a synergistic antinociceptive effect between opioidergic and dopaminergic drugs. This combination may relieve painful conditions resistant to conventional treatments, and it may reduce the adverse effects of chronic opioid administration.
Collapse
Affiliation(s)
- Jonathan Mercado-Reyes
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Angélica Almanza
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Pedro Segura-Chama
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico; Cátedras CONACyT - Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Francisco Pellicer
- Laboratorio de Neurofisiología Integrativa, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Francisco Mercado
- Laboratorio de Fisiología Celular, Dirección de Investigaciones en Neurociencias. Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico.
| |
Collapse
|
249
|
Lever JR, Fergason-Cantrell EA, Carmack TL, Watkinson LD, Gallazzi F. Design, synthesis and evaluation of 111In labeled DOTA-conjugated tetrapeptides having high affinity and selectivity for mu opioid receptors. Nucl Med Biol 2019; 70:53-66. [PMID: 30933866 DOI: 10.1016/j.nucmedbio.2019.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/10/2019] [Accepted: 02/17/2019] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Peripheral mu (μ) opioid receptors are implicated in pain, bowel dysfunction and the progression of certain cancers. In an effort to identify radioligands well suited for imaging these peripheral sites, we have prepared and evaluated four hydrophilic 111In labeled DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) conjugated μ tetrapeptides. METHODS Peptides were prepared by solid-phase techniques, using orthogonal strategies to achieve branching to DOTA, and then characterized by HPLC, mass spectroscopy and amino acid analysis. Scaffolds included novel peptide H-Dmt-D-Ala-Phe-Orn-NH2 (DAPO), where Dmt = 2',6'-dimethyltyrosine, and known peptide H-Dmt-D-Arg-Phe-Lys-NH2 ([Dmt1]DALDA). Constructs had DOTA conjugation at the Orn4 or Lys4 side chains, or to the C-terminal through a hexanoic acid-lysine linker. Indium(III) complexation and 111In radiolabeling were accomplished by standard methods. Protein binding and Log D7.4 were determined. Binding and pharmacological profiles were obtained in vitro. Biodistribution and radiometabolite studies were conducted using male CD-1 mice. RESULTS All four indium(III)-DOTA conjugates derived from DAPO and [Dmt1]DALDA showed good selectivity and subnanomolar affinity for μ opioid receptors. One radioligand, H-Dmt-D-Ala-Phe-Orn(δ-[111In]In-DOTA)-NH2, showed 25% specific binding in vivo to μ sites in mouse gut. Notably, this was the least polar of the series, and also showed low sensitivity to modulation of binding by sodium ions. All radioligands showed high kidney uptake of radiometabolites. CONCLUSIONS Visualizing peripheral μ opioid receptors using 111In labeled DOTA-conjugated tetrapeptides appears feasible, but structural modifications to enhance specific binding and metabolic stability, as well as to reduce kidney uptake, will be required. ADVANCES IN KNOWLEDGE This study shows in vivo labeling of peripheral μ opioid receptors by a tetrapeptide radioligand, and provides information that should prove useful in the design of peptide radioligands having optimal properties.
Collapse
Affiliation(s)
- John R Lever
- Department of Radiology, University of Missouri, Columbia, MO 65212, USA; Radiopharmaceutical Sciences Institute, University of Missouri, Columbia, MO 65211, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA.
| | - Emily A Fergason-Cantrell
- Department of Radiology, University of Missouri, Columbia, MO 65212, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Terry L Carmack
- Radiopharmaceutical Sciences Institute, University of Missouri, Columbia, MO 65211, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Lisa D Watkinson
- Radiopharmaceutical Sciences Institute, University of Missouri, Columbia, MO 65211, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| | - Fabio Gallazzi
- Department of Chemistry, University of Missouri, Columbia, MO 65211, USA; Molecular Interaction Core, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
250
|
Asante DB, Henneh IT, Acheampong DO, Kyei F, Adokoh CK, Ofori EG, Domey NK, Adakudugu E, Tangella LP, Ameyaw EO. Anti-inflammatory, anti-nociceptive and antipyretic activity of young and old leaves of Vernonia amygdalina. Biomed Pharmacother 2019; 111:1187-1203. [DOI: 10.1016/j.biopha.2018.12.147] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/28/2018] [Accepted: 12/31/2018] [Indexed: 12/29/2022] Open
|