1
|
Cardona-Acosta AM, Meisser N, Vardeleon NI, Steiner H, Bolaños-Guzmán CA. Mother's little helper turned a foe: Alprazolam use, misuse, and abuse. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111137. [PMID: 39260815 DOI: 10.1016/j.pnpbp.2024.111137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Benzodiazepines are effective in managing anxiety and related disorders when used properly (short-term). Their inappropriate use, however, carries significant risks, involving amnesia, rebound insomnia, rebound anxiety, depression, dependence, abuse, addiction, and an intense and exceedingly prolonged withdrawal, among other complications. Benzodiazepines also amplify the effects of opioids and, consequently, have been implicated in approximately 30 % of opioid overdose deaths. Despite their unfavorable profile, sharp increases in medical and non-medical use of benzodiazepines have been steadily reported worldwide. Alprazolam (Xanax®), a potent, short-acting benzodiazepine, is among the most prescribed and abused anxiolytics in the United States. This medication is commonly co-abused with opioids, increasing the likelihood for oversedation, overdose, and death. Notwithstanding these risks, it is surprising that research investigating how benzodiazepines, such as alprazolam, interact with opioids is severely lacking in clinical and preclinical settings. This review therefore aims to present our current knowledge of benzodiazepine use and misuse, with an emphasis on alprazolam when data is available, and particularly in populations at higher risk for developing substance use disorders. Additionally, the potential mechanism(s) surrounding tolerance, dependence and abuse liability are discussed. Despite their popularity, our understanding of how benzodiazepines and opioids interact is less than adequate. Therefore, it is now more important than ever to understand the short- and long-term consequences of benzodiazepine/alprazolam use.
Collapse
Affiliation(s)
- Astrid M Cardona-Acosta
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Noelle Meisser
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Nathan I Vardeleon
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Carlos A Bolaños-Guzmán
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
2
|
Chrószcz M, Hajto J, Misiołek K, Szumiec Ł, Ziemiańska M, Radlicka-Borysewska A, Borczyk M, Zięba M, Gołda S, Siwiec M, Ziółkowska B, Piechota M, Korostyński M, Rodriguez Parkitna J. μ-Opioid receptor transcriptional variants in the murine forebrain and spinal cord. Gene 2025; 932:148890. [PMID: 39187136 DOI: 10.1016/j.gene.2024.148890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
Oprm1, the gene encoding the μ-opioid receptor, has multiple reported transcripts, with a variable 3' region and many alternative sequences encoding the C-terminus of the protein. The functional implications of this variability remain mostly unexplored, though a recurring notion is that it could be exploited by developing selective ligands with improved clinical profiles. Here, we comprehensively examined Oprm1 transcriptional variants in the murine central nervous system, using long-read RNAseq as well as spatial and single-cell transcriptomics. The results were validated with RNAscope in situ hybridization. We found a mismatch between transcripts annotated in the mouse genome (GRCm38/mm10) and the RNA-seq results. Sequencing data indicated that the primary Oprm1 transcript has a 3' terminus located on chr10:6,860,027, which is ∼ 9.5 kilobases downstream of the longest annotated exon 4 end. Long-read sequencing confirmed that the final Oprm1 exon included a 10.2 kilobase long 3' untranslated region, and the presence of the long variant was unambiguously confirmed using RNAscope in situ hybridization in the thalamus, striatum, cortex and spinal cord. Conversely, expression of the Oprm1 reference transcript or alternative transcripts of the Oprm1 gene was absent or close to the detection limit. Thus, the primary transcript of the Oprm1 mouse gene is a variant with a long 3' untranslated region, which is homologous to the human OPRM1 primary transcript and encodes the same conserved C-terminal amino acid sequence.
Collapse
Affiliation(s)
- Magdalena Chrószcz
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Jacek Hajto
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Klaudia Misiołek
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Łukasz Szumiec
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Magdalena Ziemiańska
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Anna Radlicka-Borysewska
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Małgorzata Borczyk
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Mateusz Zięba
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Sławomir Gołda
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Marcin Siwiec
- Department of Physiology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Barbara Ziółkowska
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Marcin Piechota
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Michał Korostyński
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland.
| |
Collapse
|
3
|
Effah F, Nidadavolu P, de Gusmão Taveiros Silva NK, Wojtowicz M, Camarini R, Zimmer A, Bailey A. Differential temporal decline of cerebral oxytocin and μ-opioid receptor density during the aging process in mice. Eur J Neurosci 2024; 60:6686-6703. [PMID: 39435722 PMCID: PMC11612844 DOI: 10.1111/ejn.16578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024]
Abstract
Aging is often associated with changes in social, sexual, emotional and pain functioning, as well as with the increased prevalence of certain psychopathologies. However, the neurodevelopmental basis underpinning these age-related changes remains to be determined. Considering the key roles of oxytocin (OTR) and μ-opioid (MOPr) receptor systems in regulating social, sexual, pain, reward and emotional processing, it seems plausible that they are also implicated in age-related behavioural alterations. Although the ontogeny of both receptors has been well characterized in rodent brains from early development till adulthood, little is known concerning the neuroadaptations occurring from middle age to old age. Therefore, we mapped the neuroadaptations in OTR and MOPr in the brains of mice at those developmental endpoints. Quantitative OTR and MOPr autoradiographic binding was carried out in the brains of male mice at 2, 6, 9, 12 and 18 months of age. A significant whole brain decline in OTR density was detected between 2 and 6 months of age, with no additional decline thereafter. Interestingly, for MOPrs, the decline in density was not detected until 9 months of age. Region-specific age-related decline in OTR density was concentrated in the lateral anterior olfactory nuclei (AOL) and, for MOPr, in the AOL and the nucleus accumbens. Identifying the tipping point of these age-related variations in both receptors may assist with our understanding of the neurobiology underlining age-related changes in social, pain and emotional functioning/processing. It may also help us target interventions to specific developmental windows to abrogate certain age-related psychopathologies.
Collapse
Affiliation(s)
- Felix Effah
- Pharmacology Section, St. George's School of Health & Medical SciencesCity St George's University of London, Cranmer TerraceLondonUK
| | - Prakash Nidadavolu
- Institute of Molecular Psychiatry, Medical Faculty, University of BonnBonnGermany
| | | | - Milosz Wojtowicz
- Pharmacology Section, St. George's School of Health & Medical SciencesCity St George's University of London, Cranmer TerraceLondonUK
| | - Rosana Camarini
- Pharmacology DepartmentUniversidade de Sao PauloSão PauloBrazil
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of BonnBonnGermany
| | - Alexis Bailey
- Pharmacology Section, St. George's School of Health & Medical SciencesCity St George's University of London, Cranmer TerraceLondonUK
| |
Collapse
|
4
|
Falconnier C, Caparros-Roissard A, Decraene C, Lutz PE. Functional genomic mechanisms of opioid action and opioid use disorder: a systematic review of animal models and human studies. Mol Psychiatry 2023; 28:4568-4584. [PMID: 37723284 PMCID: PMC10914629 DOI: 10.1038/s41380-023-02238-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/20/2023]
Abstract
In the past two decades, over-prescription of opioids for pain management has driven a steep increase in opioid use disorder (OUD) and death by overdose, exerting a dramatic toll on western countries. OUD is a chronic relapsing disease associated with a lifetime struggle to control drug consumption, suggesting that opioids trigger long-lasting brain adaptations, notably through functional genomic and epigenomic mechanisms. Current understanding of these processes, however, remain scarce, and have not been previously reviewed systematically. To do so, the goal of the present work was to synthesize current knowledge on genome-wide transcriptomic and epigenetic mechanisms of opioid action, in primate and rodent species. Using a prospectively registered methodology, comprehensive literature searches were completed in PubMed, Embase, and Web of Science. Of the 2709 articles identified, 73 met our inclusion criteria and were considered for qualitative analysis. Focusing on the 5 most studied nervous system structures (nucleus accumbens, frontal cortex, whole striatum, dorsal striatum, spinal cord; 44 articles), we also conducted a quantitative analysis of differentially expressed genes, in an effort to identify a putative core transcriptional signature of opioids. Only one gene, Cdkn1a, was consistently identified in eleven studies, and globally, our results unveil surprisingly low consistency across published work, even when considering most recent single-cell approaches. Analysis of sources of variability detected significant contributions from species, brain structure, duration of opioid exposure, strain, time-point of analysis, and batch effects, but not type of opioid. To go beyond those limitations, we leveraged threshold-free methods to illustrate how genome-wide comparisons may generate new findings and hypotheses. Finally, we discuss current methodological development in the field, and their implication for future research and, ultimately, better care.
Collapse
Affiliation(s)
- Camille Falconnier
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, 67000, Strasbourg, France
| | - Alba Caparros-Roissard
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, 67000, Strasbourg, France
| | - Charles Decraene
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, 67000, Strasbourg, France
- Centre National de la Recherche Scientifique, Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives UMR 7364, 67000, Strasbourg, France
| | - Pierre-Eric Lutz
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, 67000, Strasbourg, France.
- Douglas Mental Health University Institute, Montreal, QC, Canada.
| |
Collapse
|
5
|
Leon Duque MA, Vallavoju N, Woo CM. Chemical tools for the opioids. Mol Cell Neurosci 2023; 125:103845. [PMID: 36948231 PMCID: PMC10247539 DOI: 10.1016/j.mcn.2023.103845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023] Open
Abstract
The opioids are potent and widely used pain management medicines despite also possessing severe liabilities that have fueled the opioid crisis. The pharmacological properties of the opioids primarily derive from agonism or antagonism of the opioid receptors, but additional effects may arise from specific compounds, opioid receptors, or independent targets. The study of the opioids, their receptors, and the development of remediation strategies has benefitted from derivatization of the opioids as chemical tools. While these studies have primarily focused on the opioids in the context of the opioid receptors, these chemical tools may also play a role in delineating mechanisms that are independent of the opioid receptors. In this review, we describe recent advances in the development and applications of opioid derivatives as chemical tools and highlight opportunities for the future.
Collapse
Affiliation(s)
- Mark Anthony Leon Duque
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA 02138, United States of America
| | - Nandini Vallavoju
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA 02138, United States of America
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA 02138, United States of America.
| |
Collapse
|
6
|
Yang C, Gong Z, Zhang X, Miao S, Li B, Xie W, Wang T, Han X, Wang L, Dong Z, Yu S. Neuropeptide Y in the medial habenula alleviates migraine-like behaviors through the Y1 receptor. J Headache Pain 2023; 24:61. [PMID: 37231359 DOI: 10.1186/s10194-023-01596-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Migraine is a highly disabling health burden with multiple symptoms; however, it remains undertreated because of an inadequate understanding of its neural mechanisms. Neuropeptide Y (NPY) has been demonstrated to be involved in the modulation of pain and emotion, and may play a role in migraine pathophysiology. Changes in NPY levels have been found in patients with migraine, but whether and how these changes contribute to migraine is unknown. Therefore, the purpose of this study was to investigate the role of NPY in migraine-like phenotypes. METHODS Here, we used intraperitoneal injection of glyceryl trinitrate (GTN, 10 mg/kg) as a migraine mouse model, which was verified by light-aversive test, von Frey test, and elevated plus maze test. We then performed whole-brain imaging with NPY-GFP mice to explore the critical regions where NPY was changed by GTN treatment. Next, we microinjected NPY into the medial habenula (MHb), and further infused Y1 or Y2 receptor agonists into the MHb, respectively, to detect the effects of NPY in GTN-induced migraine-like behaviors. RESULTS GTN effectively triggered allodynia, photophobia, and anxiety-like behaviors in mice. After that, we found a decreased level of GFP+ cells in the MHb of GTN-treated mice. Microinjection of NPY attenuated GTN-induced allodynia and anxiety without affecting photophobia. Furthermore, we found that activation of Y1-but not Y2-receptors attenuated GTN-induced allodynia and anxiety. CONCLUSIONS Taken together, our data support that the NPY signaling in the MHb produces analgesic and anxiolytic effects through the Y1 receptor. These findings may provide new insights into novel therapeutic targets for the treatment of migraine.
Collapse
Affiliation(s)
- Chunxiao Yang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Zihua Gong
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Medical Oncology, 980th Hospital of PLA Joint Logistical Support Force (Bethune International Peace Hospital), Shijiazhuang, Hebei, 050082, China
| | - Xiaochen Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Shuai Miao
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Bozhi Li
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Wei Xie
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Tao Wang
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Xun Han
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
| | - Liang Wang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Zhao Dong
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Shengyuan Yu
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853, China.
- Medical School of Chinese PLA, Beijing, 100853, China.
| |
Collapse
|
7
|
Levinstein MR, Ventriglia EN, Gomez JL, Budinich RC, Marton J, Henriksen G, Holt DP, Dannals RF, Pomper MG, Zarate CA, Bonaventura J, Michaelides M. 6-O-(2-[ 18F]Fluoroethyl)-6-O-Desmethyl-Diprenorphine ([ 18F]FE-DPN) Preferentially Binds to Mu Opioid Receptors In Vivo. Mol Imaging Biol 2023; 25:384-390. [PMID: 35999424 DOI: 10.1007/s11307-022-01767-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE 6-O-(2-[18F]Fluoroethyl)-6-O-desmethyl-diprenorphine ([18F]FE-DPN) is regarded as a non-selective opioid receptor radiotracer. PROCEDURE Here, we report the first characterization of [18F]FE-DPN synthesized from the novel precursor, 6-O-(2-tosyloxyethoxy)-6-O-desmethyl-3-O-trityl-diprenorphine (TE-TDDPN), using a one-pot, two-step nucleophilic radiosynthesis to image opioid receptors in rats and mice using positron emission tomography. RESULTS We also show that [18F]FE-DPN and [3H]DPN exhibit negligible brain uptake in mu opioid receptor (MOR) knockout mice. CONCLUSIONS Taken together with prior findings, our results suggest that [18F]FE-DPN and [3H]DPN preferentially bind to MOR in rodents in vivo.
Collapse
Affiliation(s)
- Marjorie R Levinstein
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute On Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Emilya N Ventriglia
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute On Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute On Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - Reece C Budinich
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute On Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA
| | - János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Strasse 10-14, 01454, Radeberg, Germany
| | - Gjermund Henriksen
- Institute of Basic Medical Sciences, University of Oslo, Blindern, P. O. Box 1105, N-0317, Oslo, Norway.,Norwegian Medical Cyclotron Centre Ltd, Sognsvannsveien 20, N-0372, Oslo, Norway.,Institute of Physics, University of Oslo, Sem Sælands vei 24, N-0371, Oslo, Norway
| | - Daniel P Holt
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Robert F Dannals
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Martin G Pomper
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, Bethesda, MD, 20892, USA
| | - Jordi Bonaventura
- Departament de Patologia I Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalonia, Spain.,Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907, L'Hospitalet de Llobregat, Catalonia, Spain
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute On Drug Abuse Intramural Research Program, Baltimore, MD, 21224, USA. .,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
8
|
Dynamic Changes in the Endocannabinoid System during the Aging Process: Focus on the Middle-Age Crisis. Int J Mol Sci 2022; 23:ijms231810254. [PMID: 36142165 PMCID: PMC9499672 DOI: 10.3390/ijms231810254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Endocannabinoid (eCB) signaling is markedly decreased in the hippocampus (Hip) of aged mice, and the genetic deletion of the cannabinoid receptor type 1 (CB1) leads to an early onset of cognitive decline and age-related histological changes in the brain. Thus, it is hypothesized that cognitive aging is modulated by eCB signaling through CB1. In the present study, we detailed the changes in the eCB system during the aging process using different complementary techniques in mouse brains of five different age groups, ranging from adolescence to old age. Our findings indicate that the eCB system is most strongly affected in middle-aged mice (between 9 and 12 months of age) in a brain region-specific manner. We show that 2-arachidonoylglycerol (2-AG) was prominently decreased in the Hip and moderately in caudate putamen (CPu), whereas anandamide (AEA) was decreased in both CPu and medial prefrontal cortex along with cingulate cortex (mPFC+Cg), starting from 6 months until 12 months. Consistent with the changes in 2-AG, the 2-AG synthesizing enzyme diacylglycerol lipase α (DAGLα) was also prominently decreased across the sub-regions of the Hip. Interestingly, we found a transient increase in CB1 immunoreactivity across the sub-regions of the Hip at 9 months, a plausible compensation for reduced 2-AG, which ultimately decreased strongly at 12 months. Furthermore, quantitative autoradiography of CB1 revealed that [3H]CP55940 binding markedly increased in the Hip at 9 months. However, unlike the protein levels, CB1 binding density did not drop strongly at 12 months and at old age. Furthermore, [3H]CP55940 binding was significantly increased in the lateral entorhinal cortex (LEnt), starting from the middle age until the old age. Altogether, our findings clearly indicate a middle-age crisis in the eCB system, which could be a potential time window for therapeutic interventions to abrogate the course of cognitive aging.
Collapse
|
9
|
Effah F, de Gusmão Taveiros Silva NK, Vijayanathan K, Camarini R, Joly F, Taiwo B, Rabot S, Champeil-Potokar G, Bombail V, Bailey A. SEX-DEPENDENT IMPACT OF MICROBIOTA STATUS ON CEREBRAL μ -OPIOID RECEPTOR DENSITY IN FISCHER RATS. Eur J Neurosci 2022; 55:1917-1933. [PMID: 35393704 PMCID: PMC9324823 DOI: 10.1111/ejn.15666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/08/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022]
Abstract
μ‐opioid receptors (MOPr) play a critical role in social play, reward and pain, in a sex‐ and age‐dependent manner. There is evidence to suggest that sex and age differences in brain MOPr density may be responsible for this variability; however, little is known about the factors driving these differences in cerebral MOPr density. Emerging evidence highlights gut microbiota's critical influence and its bidirectional interaction with the brain on neurodevelopment. Therefore, we aimed to determine the impact of gut microbiota on MOPr density in male and female brains at different developmental stages. Quantitative [3H]DAMGO autoradiographic binding was carried out in the forebrain of male and female conventional (CON) and germ‐free (GF) rats at postnatal days (PND) 8, 22 and 116–150. Significant ‘microbiota status X sex’, ‘age X brain region’ interactions and microbiota status‐ and age‐dependent effects on MOPr binding were uncovered. Microbiota status influenced MOPr levels in males but not females, with higher MOPr levels observed in GF versus CON rats overall regions and age groups. In contrast, no overall sex differences were observed in GF or CON rats. Interestingly, within‐age planned comparison analysis conducted in frontal cortical and brain regions associated with reward revealed that this microbiota effect was restricted only to PND22 rats. Thus, this pilot study uncovers the critical sex‐dependent role of gut microbiota in regulating cerebral MOPr density, which is restricted to the sensitive developmental period of weaning. This may have implications in understanding the importance of microbiota during early development on opioid signalling and associated behaviours.
Collapse
Affiliation(s)
- Felix Effah
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | | | - Katie Vijayanathan
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | - Rosana Camarini
- Pharmacology Department, Universidade de Sao Paulo, São Paulo, Brazil
| | - Fatima Joly
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Benjamin Taiwo
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Vincent Bombail
- UMR PNCA, AgroParisTech, INRAE, Université Paris-Saclay, Paris, France
| | - Alexis Bailey
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| |
Collapse
|
10
|
Osman A, Zuffa S, Walton G, Fagbodun E, Zanos P, Georgiou P, Kitchen I, Swann J, Bailey A. Post-weaning A1/A2 β-casein milk intake modulates depressive-like behavior, brain μ-opioid receptors, and the metabolome of rats. iScience 2021; 24:103048. [PMID: 34585111 PMCID: PMC8450247 DOI: 10.1016/j.isci.2021.103048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/13/2021] [Accepted: 08/24/2021] [Indexed: 02/05/2023] Open
Abstract
The postnatal period is critical for brain and behavioral development and is sensitive to environmental stimuli, such as nutrition. Prevention of weaning from maternal milk was previously shown to cause depressive-like behavior in rats. Additionally, loss of dietary casein was found to act as a developmental trigger for a population of brain opioid receptors. Here, we explore the effect of exposure to milk containing A1 and A2 β-casein beyond weaning. A1 but not A2 β-casein milk significantly increased stress-induced immobility in rats, concomitant with an increased abundance of Clostridium histolyticum bacterial group in the caecum and colon of A1 β-casein fed animals, brain region-specific alterations of μ-opioid and oxytocin receptors, and modifications in urinary biochemical profiles. Moreover, urinary gut microbial metabolites strongly correlated with altered brain metabolites. These findings suggest that consumption of milk containing A1 β-casein beyond weaning age may affect mood via a possible gut-brain axis mechanism. Postnatal brain development is sensitive to nutritional exposures Consumption of A1 but not A2 β-casein milk post-weaning affects mood in rats Gut microbial, biochemical, and neurochemical changes accompany mood alterations Urinary gut microbial metabolites correlate with brain metabolites
Collapse
Affiliation(s)
- Aya Osman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Simone Zuffa
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Gemma Walton
- Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Reading, UK
| | - Elizabeth Fagbodun
- Pharmacology Section, Institute of Medical and Biomedical Education, St George's University of London, London, UK
| | - Panos Zanos
- Department of Psychology, University of Cyprus, 1 University Avenue, 2109 Nicosia, Cyprus
| | - Polymnia Georgiou
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Ian Kitchen
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | - Jonathan Swann
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK.,School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Alexis Bailey
- Pharmacology Section, Institute of Medical and Biomedical Education, St George's University of London, London, UK
| |
Collapse
|
11
|
Addiction and the cerebellum with a focus on actions of opioid receptors. Neurosci Biobehav Rev 2021; 131:229-247. [PMID: 34555385 DOI: 10.1016/j.neubiorev.2021.09.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/12/2021] [Accepted: 09/12/2021] [Indexed: 01/19/2023]
Abstract
Increasing evidence suggests that the cerebellum could play a role in the higher cognitive processes involved in addiction as the cerebellum contains anatomical and functional pathways to circuitry controlling motivation and saliency. In addition, the cerebellum exhibits a widespread presence of receptors, including opioid receptors which are known to play a prominent role in synaptic and circuit mechanisms of plasticity associated with drug use and development of addiction to opioids and other drugs of abuse. Further, the presence of perineural nets (PNNs) in the cerebellum which contain proteins known to alter synaptic plasticity could contribute to addiction. The role the cerebellum plays in processes of addiction is likely complex, and could depend on the particular drug of abuse, the pattern of use, and the stage of the user within the addiction cycle. In this review, we discuss functional and structural modifications shown to be produced in the cerebellum by opioids that exhibit dependency-inducing properties which provide support for the conclusion that the cerebellum plays a role in addiction.
Collapse
|
12
|
Fritzwanker S, Moulédous L, Mollereau C, Froment C, Burlet-Schiltz O, Effah F, Bailey A, Spetea M, Reinscheid RK, Schulz S, Kliewer A. HA-MOP knockin mice express the canonical µ-opioid receptor but lack detectable splice variants. Commun Biol 2021; 4:1070. [PMID: 34522000 PMCID: PMC8440528 DOI: 10.1038/s42003-021-02580-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/17/2021] [Indexed: 12/31/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are notoriously difficult to detect in native tissues. In an effort to resolve this problem, we have developed a novel mouse model by fusing the hemagglutinin (HA)-epitope tag sequence to the amino-terminus of the µ-opioid receptor (MOP). Although HA-MOP knock-in mice exhibit reduced receptor expression, we found that this approach allowed for highly efficient immunodetection of low abundant GPCR targets. We also show that the HA-tag facilitates both high-resolution imaging and immunoisolation of MOP. Mass spectrometry (MS) confirmed post-translational modifications, most notably agonist-selective phosphorylation of carboxyl-terminal serine and threonine residues. MS also unequivocally identified the carboxyl-terminal 387LENLEAETAPLP398 motif, which is part of the canonical MOP sequence. Unexpectedly, MS analysis of brain lysates failed to detect any of the 15 MOP isoforms that have been proposed to arise from alternative splicing of the MOP carboxyl-terminus. For quantitative analysis, we performed multiple successive rounds of immunodepletion using the well-characterized rabbit monoclonal antibody UMB-3 that selectively detects the 387LENLEAETAPLP398 motif. We found that >98% of HA-tagged MOP contain the UMB-3 epitope indicating that virtually all MOP expressed in the mouse brain exhibit the canonical amino acid sequence.
Collapse
Affiliation(s)
- Sebastian Fritzwanker
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Lionel Moulédous
- Research Center on Animal Cognition, Center for Integrative Biology, CNRS, UPS, Toulouse University, Toulouse, France
| | - Catherine Mollereau
- Research Center on Animal Cognition, Center for Integrative Biology, CNRS, UPS, Toulouse University, Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Carine Froment
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Felix Effah
- Pharmacology Section, St George's University of London, London, UK
| | - Alexis Bailey
- Pharmacology Section, St George's University of London, London, UK
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Rainer K Reinscheid
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Stefan Schulz
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany.
| | - Andrea Kliewer
- Department of Pharmacology and Toxicology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
13
|
Arttamangkul S, Platt EJ, Carroll J, Farrens D. Functional independence of endogenous µ- and δ-opioid receptors co-expressed in cholinergic interneurons. eLife 2021; 10:69740. [PMID: 34477106 PMCID: PMC8718112 DOI: 10.7554/elife.69740] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/31/2021] [Indexed: 12/18/2022] Open
Abstract
Class A G-protein-coupled receptors (GPCRs) normally function as monomers, although evidence from heterologous expression systems suggests that they may sometimes form homodimers and/or heterodimers. This study aims to evaluate possible functional interplay of endogenous µ- and δ-opioid receptors (MORs and DORs) in mouse neurons. Detecting GPCR dimers in native tissues, however, has been challenging. Previously, MORs and DORs co-expressed in transfected cells have been reported to form heterodimers, and their possible co-localization in neurons has been studied in knock-in mice expressing genetically engineered receptors fused to fluorescent proteins. Here, we find that single cholinergic neurons in the mouse striatum endogenously express both MORs and DORs. The receptors on neurons from live brain slices were fluorescently labeled with a ligand-directed labeling reagent, NAI-A594. The selective activation of MORs and DORs, with DAMGO (µ-agonist) and deltorphin (δ-agonist) inhibited spontaneous firing in all cells examined. In the continued presence of agonist, the firing rate returned to baseline as the result of receptor desensitization with the application of deltorphin but was less observed with the application of DAMGO. In addition, agonist-induced internalization of DORs but not MORs was detected. When MORs and DORs were activated simultaneously with [Met5]-enkephalin, desensitization of MORs was facilitated but internalization was not increased. Together, these results indicate that while MORs and DORs are expressed in single striatal cholinergic interneurons, the two receptors function independently.
Collapse
Affiliation(s)
| | - Emily J Platt
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, United States
| | - James Carroll
- Surgery, Oregon Health and Science University, Portland, United States
| | - David Farrens
- Department of Biochemistry and Molecular Biology, School of Medicine, Oregon Health and Science University, Portland, United States
| |
Collapse
|
14
|
Weiss N, Zamponi GW. Opioid Receptor Regulation of Neuronal Voltage-Gated Calcium Channels. Cell Mol Neurobiol 2021; 41:839-847. [PMID: 32514826 DOI: 10.1007/s10571-020-00894-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/28/2022]
Abstract
Neuronal voltage-gated calcium channels play a pivotal role in the conversion of electrical signals into calcium entry into nerve endings that is required for the release of neurotransmitters. They are under the control of a number of cellular signaling pathways that serve to fine tune synaptic activities, including G-protein coupled receptors (GPCRs) and the opioid system. Besides modulating channel activity via activation of second messengers, GPCRs also physically associate with calcium channels to regulate their function and expression at the plasma membrane. In this mini review, we discuss the mechanisms by which calcium channels are regulated by classical opioid and nociceptin receptors. We highlight the importance of this regulation in the control of neuronal functions and their implication in the development of disease conditions. Finally, we present recent literature concerning the use of novel μ-opioid receptor/nociceptin receptor modulators and discuss their use as potential drug candidates for the treatment of pain.
Collapse
Affiliation(s)
- Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
15
|
Saleh AH, Abdelwaly A, Darwish KM, Eissa AAHM, Chittiboyina A, Helal MA. Deciphering the molecular basis of the kappa opioid receptor selectivity: A Molecular Dynamics study. J Mol Graph Model 2021; 106:107940. [PMID: 34015577 DOI: 10.1016/j.jmgm.2021.107940] [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: 03/05/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/30/2022]
Abstract
Selective antagonists for the kappa opioid receptor (KOP) have the potential to treat opiate and alcohol addiction, as well as depression and other CNS disorders. Over the years, the development of KOP-selective antagonists yielded very few successful compounds. Recently, N-substituted trans-3,4-dimethyl-4-(3-hydroxyphenyl)piperidines have emerged as a novel class of pure opioid receptor antagonists, including the marketed Mu opioid receptor (MOP) peripheral antagonist Alvimopan and the potent KOP antagonist JDTic. However, the selectivity determinants of this class of compounds towards the opioid receptor subtypes are still vague and understudied. In this work, we have performed Molecular Dynamics (MD) simulation to gain insights into the differential binding of this class of compounds into KOP, as exemplified by Alvimopan and JDTic. Our study indicated that the selectivity largely depends on ligands interaction with the selectivity pocket formed by Val108, Thr111, and Val118, supported by two additional polar and hydrophobic contacts with Asp138 and Trp287, respectively. Our results also demonstrate, for the first time, that non-morphinan ligands can still adopt the "message-address model" for KOP efficacy and selectivity by binding to Glu297.
Collapse
Affiliation(s)
- Amr H Saleh
- University of Science and Technology, Biomedical Sciences Program, Zewail City of Science and Technology, October Gardens, 6th of October, Giza, 12578, Egypt
| | - Ahmad Abdelwaly
- University of Science and Technology, Biomedical Sciences Program, Zewail City of Science and Technology, October Gardens, 6th of October, Giza, 12578, Egypt
| | - Khaled M Darwish
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Amal A H M Eissa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Amar Chittiboyina
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS, 38677, United States
| | - Mohamed A Helal
- University of Science and Technology, Biomedical Sciences Program, Zewail City of Science and Technology, October Gardens, 6th of October, Giza, 12578, Egypt; Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
16
|
Effah F, de Gusmão Taveiros Silva NK, Camarini R, Joly F, Rabot S, Bombail V, Bailey A. Region-specific sex modulation of central oxytocin receptor by gut microbiota: An ontogenic study. Dev Neurobiol 2021; 81:149-163. [PMID: 33389811 DOI: 10.1002/dneu.22805] [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: 06/22/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 01/14/2023]
Abstract
Oxytocin (OT) is a developmentally important neuropeptide recognized to play a dominant role in social functioning and stress-related behaviors, in a sex-dependent manner. Nonetheless, the underlining factors driving OT and OT receptor (OTR) early brain development remain unclear. Recent evidence highlight the critical influence of gut microbiota and its bidirectional interaction with the brain on neurodevelopment via the gut microbiota-brain axis. Therefore, we aimed to determine the impact of gut microbiota on the OTR system of the rat brain at different developmental stages in a pilot study. Quantitative OTR [125 I]-OVTA autoradiographic binding was carried out in the forebrain of male and female conventional (CON) and germ-free (GF) rats at postnatal days (PND) 8, 22, and 116-150. OTR binding was also assessed in the eyes of PND 1 and PND 4 GF female rats. Significant "microbiota × sex × region" interaction and age-dependent effects on OTR binding were demonstrated. Microbiota status influenced OTR levels in males but not females with higher levels of OTR observed in GF versus CON rats in the cingulate, prelimbic, and lateral/medial/ventral orbital cortex, and septum across all age groups, while sex differences were observed in GF, but not in CON rats. Interestingly, OTRs present in the eyes of CON rats were abolished in GF rats. This is the first study to uncover a sex-specific role of gut microbiota on the central OTR system, which may have implications in understanding the developmental neuroadaptations critical for behavioral regulation and the etiology of certain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Felix Effah
- Pharmacology Section, St George's University of London, London, UK
| | | | - Rosana Camarini
- Pharmacology Department, Universidade de Sao Paulo, São Paulo, Brazil
| | - Fatima Joly
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Sylvie Rabot
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
| | - Vincent Bombail
- UMR PNCA, AgroParisTech, INRAE, Université Paris-Saclay, Paris, France
| | - Alexis Bailey
- Pharmacology Section, St George's University of London, London, UK
| |
Collapse
|
17
|
Galaj E, Xi ZX. Progress in opioid reward research: From a canonical two-neuron hypothesis to two neural circuits. Pharmacol Biochem Behav 2021; 200:173072. [PMID: 33227308 PMCID: PMC7796909 DOI: 10.1016/j.pbb.2020.173072] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/21/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022]
Abstract
Opioid abuse and related overdose deaths continue to rise in the United States, contributing to the national opioid crisis in the USA. The neural mechanisms underlying opioid abuse and addiction are still not fully understood. This review discusses recent progress in basic research dissecting receptor mechanisms and circuitries underlying opioid reward and addiction. We first review the canonical GABA-dopamine neuron hypothesis that was upheld for half a century, followed by major findings challenging this hypothesis. We then focus on recent progress in research evaluating the role of the mesolimbic and nigrostriatal dopamine circuitries in opioid reward and relapse. Based on recent findings that activation of dopamine neurons in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) is equally rewarding and that GABA neurons in the rostromedial tegmental nucleus (RMTg) and the substantia nigra pars reticula (SNr) are rich in mu opioid receptors and directly synapse onto midbrain DA neurons, we proposed that the RTMg→VTA → ventrostriatal and SNr → SNc → dorsostriatal pathways may act as the two major neural substrates underlying opioid reward and abuse. Lastly, we discuss possible integrations of these two pathways during initial opioid use, development of opioid abuse and maintenance of compulsive opioid seeking.
Collapse
Affiliation(s)
- Ewa Galaj
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States of America
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States of America.
| |
Collapse
|
18
|
Derouiche L, Pierre F, Doridot S, Ory S, Massotte D. Heteromerization of Endogenous Mu and Delta Opioid Receptors Induces Ligand-Selective Co-Targeting to Lysosomes. Molecules 2020; 25:molecules25194493. [PMID: 33007971 PMCID: PMC7583997 DOI: 10.3390/molecules25194493] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/23/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence indicates that native mu and delta opioid receptors can associate to form heteromers in discrete brain neuronal circuits. However, little is known about their signaling and trafficking. Using double-fluorescent knock-in mice, we investigated the impact of neuronal co-expression on the internalization profile of mu and delta opioid receptors in primary hippocampal cultures. We established ligand selective mu–delta co-internalization upon activation by 1-[[4-(acetylamino)phenyl]methyl]-4-(2-phenylethyl)-4-piperidinecarboxylic acid, ethyl ester (CYM51010), [d-Ala2, NMe-Phe4, Gly-ol5]enkephalin (DAMGO), and deltorphin II, but not (+)-4-[(αR)-α-((2S,5R)-4-Allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80), morphine, or methadone. Co-internalization was driven by the delta opioid receptor, required an active conformation of both receptors, and led to sorting to the lysosomal compartment. Altogether, our data indicate that mu–delta co-expression, likely through heteromerization, alters the intracellular fate of the mu opioid receptor, which provides a way to fine-tune mu opioid receptor signaling. It also represents an interesting emerging concept for the development of novel therapeutic drugs and strategies.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Endocytosis
- Hippocampus/cytology
- Ligands
- Lysosomes
- Mice, Inbred C57BL
- Neurons/metabolism
- Piperidines/pharmacology
- Protein Multimerization
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Lyes Derouiche
- French National Centre for Scientific Research, Institut des Neurosciences Cellulaires et Intégratives, University of Strasbourg, 67000 Strasbourg, France; (L.D.); (F.P.); (S.O.)
| | - Florian Pierre
- French National Centre for Scientific Research, Institut des Neurosciences Cellulaires et Intégratives, University of Strasbourg, 67000 Strasbourg, France; (L.D.); (F.P.); (S.O.)
| | - Stéphane Doridot
- French National Centre for Scientific Research, Chronobiotron, 67200 Strasbourg, France;
| | - Stéphane Ory
- French National Centre for Scientific Research, Institut des Neurosciences Cellulaires et Intégratives, University of Strasbourg, 67000 Strasbourg, France; (L.D.); (F.P.); (S.O.)
| | - Dominique Massotte
- French National Centre for Scientific Research, Institut des Neurosciences Cellulaires et Intégratives, University of Strasbourg, 67000 Strasbourg, France; (L.D.); (F.P.); (S.O.)
- Correspondence:
| |
Collapse
|
19
|
Galaj E, Newman AH, Xi ZX. Dopamine D3 receptor-based medication development for the treatment of opioid use disorder: Rationale, progress, and challenges. Neurosci Biobehav Rev 2020; 114:38-52. [PMID: 32376243 PMCID: PMC7252042 DOI: 10.1016/j.neubiorev.2020.04.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 01/11/2023]
Abstract
Opioid abuse and related overdose deaths continue to rise in the United States, contributing to the current national opioid crisis. Although several opioid-based pharmacotherapies are available (e.g., methadone, buprenorphine, naloxone), they show limited effectiveness in long-term relapse prevention. In response to the opioid crisis, the National Institute on Drug Abuse proposed a list of pharmacological targets of highest priority for medication development for the treatment of opioid use disorders (OUD). Among these are antagonists of dopamine D3 receptors (D3R). In this review, we first review recent progress in research of the dopamine hypothesis of opioid reward and abuse and then describe the rationale and recent development of D3R ligands for the treatment of OUD. Herein, an emphasis is placed on the effectiveness of newly developed D3R antagonists in the animal models of OUD. These new drug candidates may also potentiate the analgesic effects of clinically used opioids, making them attractive as adjunctive medications for pain management and treatment of OUD.
Collapse
Affiliation(s)
- Ewa Galaj
- Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Amy Hauck Newman
- Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Zheng-Xiong Xi
- Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States.
| |
Collapse
|
20
|
Agonist-induced phosphorylation bar code and differential post-activation signaling of the delta opioid receptor revealed by phosphosite-specific antibodies. Sci Rep 2020; 10:8585. [PMID: 32444688 PMCID: PMC7244497 DOI: 10.1038/s41598-020-65589-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/05/2020] [Indexed: 01/08/2023] Open
Abstract
The δ-opioid receptor (DOP) is an attractive pharmacological target due to its potent analgesic, anxiolytic and anti-depressant activity in chronic pain models. However, some but not all selective DOP agonists also produce severe adverse effects such as seizures. Thus, the development of novel agonists requires a profound understanding of their effects on DOP phosphorylation, post-activation signaling and dephosphorylation. Here we show that agonist-induced DOP phosphorylation at threonine 361 (T361) and serine 363 (S363) proceeds with a temporal hierarchy, with S363 as primary site of phosphorylation. This phosphorylation is mediated by G protein-coupled receptor kinases 2 and 3 (GRK2/3) followed by DOP endocytosis and desensitization. DOP dephosphorylation occurs within minutes and is predominantly mediated by protein phosphatases (PP) 1α and 1β. A comparison of structurally diverse DOP agonists and clinically used opioids demonstrated high correlation between G protein-dependent signaling efficacies and receptor internalization. In vivo, DOP agonists induce receptor phosphorylation in a dose-dependent and agonist-selective manner that could be blocked by naltrexone in DOP-eGFP mice. Together, our studies provide novel tools and insights for ligand-activated DOP signaling in vitro and in vivo and suggest that DOP agonist efficacies may determine receptor post-activation signaling.
Collapse
|
21
|
Sivalingam M, Ogawa S, Parhar IS. Mapping of Morphine-Induced OPRM1 Gene Expression Pattern in the Adult Zebrafish Brain. Front Neuroanat 2020; 14:5. [PMID: 32153369 PMCID: PMC7044135 DOI: 10.3389/fnana.2020.00005] [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: 09/23/2019] [Accepted: 01/29/2020] [Indexed: 11/13/2022] Open
Abstract
Morphine is a potent analgesic opiate commonly used in treating pain, and it is also a substance of abuse and highly addictive. Hence, it is vital to discover the action sites of morphine in the brain to increase its efficacy of treatment. In the present study, we aimed at identifying comprehensive neuroanatomical locations that are sensitive to morphine in the adult zebrafish (Danio rerio). We performed in situ hybridization to localize the mu opioid receptor (oprm1) gene and to map the morphine sensitive brain areas using neuronal PAS domain-containing protein 4a (npas4a), an early gene marker. Real-time PCR was used to detect changes in mRNA levels of oprm1 and npas4a in control and acute morphine treated fish (2 mg/L; 20 min). Intense positive oprm1 signals were seen in the telencephalon, preoptic area, habenula, hypothalamic area and periventricular gray zone of the optic tectum. Acute morphine exposure significantly increased oprm1 and npas4a mRNA levels in the medial zone of dorsal telencephalon (Dm), ventral region of the ventral telencephalon (Vv), preoptic area, and in the hypothalamus but a decrease in oprm1 and npas4a signals in the dorsal habenula. This study provides a detailed map of oprm1 localization in the brain, which includes previously unreported oprm1 in the habenula of teleost. Presence of oprm1 in multiple brain sites implies multiple action targets of morphine and potential brain functions which could include reward, cognitive and negative emotions.
Collapse
Affiliation(s)
- Mageswary Sivalingam
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Satoshi Ogawa
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ishwar S Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
22
|
Wright VL, Georgiou P, Bailey A, Heal DJ, Bailey CP, Wonnacott S. Inhibition of alpha7 nicotinic receptors in the ventral hippocampus selectively attenuates reinstatement of morphine-conditioned place preference and associated changes in AMPA receptor binding. Addict Biol 2019; 24:590-603. [PMID: 29667304 PMCID: PMC6563460 DOI: 10.1111/adb.12624] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/31/2018] [Accepted: 03/02/2018] [Indexed: 01/10/2023]
Abstract
Recurrent relapse is a major problem in treating opiate addiction. Pavlovian conditioning plays a role in recurrent relapse whereby exposure to cues learned during drug intake can precipitate relapse to drug taking. α7 nicotinic acetylcholine receptors (nAChRs) have been implicated in attentional aspects of cognition and mechanisms of learning and memory. In this study we have investigated the role of α7 nAChRs in morphine-conditioned place preference (morphine-CPP). CPP provides a model of associative learning that is pertinent to associative aspects of drug dependence. The α7 nAChR antagonist methyllycaconitine (MLA; 4 mg/kg s.c.) had no effect on the acquisition, maintenance, reconsolidation or extinction of morphine-CPP but selectively attenuated morphine-primed reinstatement of CPP, in both mice and rats. Reinstatement of morphine-CPP in mice was accompanied by a selective increase in [3 H]-AMPA binding (but not in [3 H]-MK801 binding) in the ventral hippocampus that was prevented by prior treatment with MLA. Administration of MLA (6.7 μg) directly into the ventral hippocampus of rats prior to a systemic priming dose of morphine abolished reinstatement of morphine-CPP, whereas MLA delivered into the dorsal hippocampus or prefrontal cortex was without effect. These results suggest that α7 nAChRs in the ventral hippocampus play a specific role in the retrieval of associative drug memories following a period of extinction, making them potential targets for the prevention of relapse.
Collapse
Affiliation(s)
| | - Polymnia Georgiou
- Faculty of Health and Medical SciencesUniversity of SurreyUK
- Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Alexis Bailey
- Faculty of Health and Medical SciencesUniversity of SurreyUK
- Institute of Medical and Biomedical EducationSt George's University of LondonUK
| | | | | | | |
Collapse
|
23
|
Pierre F, Ugur M, Faivre F, Doridot S, Veinante P, Massotte D. Morphine-dependent and abstinent mice are characterized by a broader distribution of the neurons co-expressing mu and delta opioid receptors. Neuropharmacology 2019; 152:30-41. [PMID: 30858104 DOI: 10.1016/j.neuropharm.2019.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/04/2019] [Indexed: 02/05/2023]
Abstract
Opiate addiction develops as a chronic relapsing disorder upon drug recreational use or following misuse of analgesic prescription. Mu opioid (MOP) receptors are the primary molecular target of opiates but increasing evidence support in vivo functional heteromerization with the delta opioid (DOP) receptor, which may be part of the neurobiological processes underlying opiate addiction. Here, we used double knock-in mice co-expressing fluorescent versions of the MOP and DOP receptors to examine the impact of chronic morphine administration on the distribution of neurons co-expressing the two receptors. Our data show that MOP/DOP neuronal co-expression is broader in morphine-dependent mice and is detected in novel brain areas located in circuits related to drug reward, motor activity, visceral control and emotional processing underlying withdrawal. After four weeks of abstinence, MOP/DOP neuronal co-expression is still detectable in a large number of these brain areas except in the motor circuit. Importantly, chronic morphine administration increased the proportion of MOP/DOP neurons in the brainstem of morphine-dependent and abstinent mice. These findings establish persistent changes in the abstinent state that may modulate relapse and opiate-induced hyperalgesia and also point to the therapeutic potential of MOP/DOP targeting. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Florian Pierre
- Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Muzeyyen Ugur
- Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Fanny Faivre
- Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Stéphane Doridot
- Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France; Centre National de la Recherche Scientifique, Chronobiotron UMS 3415, Strasbourg, France
| | - Pierre Veinante
- Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Dominique Massotte
- Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France.
| |
Collapse
|
24
|
Smith JB, Alloway KD, Hof PR, Orman R, Reser DH, Watakabe A, Watson GDR. The relationship between the claustrum and endopiriform nucleus: A perspective towards consensus on cross-species homology. J Comp Neurol 2019; 527:476-499. [PMID: 30225888 PMCID: PMC6421118 DOI: 10.1002/cne.24537] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 01/08/2023]
Abstract
With the emergence of interest in studying the claustrum, a recent special issue of the Journal of Comparative Neurology dedicated to the claustrum (Volume 525, Issue 6, pp. 1313-1513) brought to light questions concerning the relationship between the claustrum (CLA) and a region immediately ventral known as the endopiriform nucleus (En). These structures have been identified as separate entities in rodents but appear as a single continuous structure in primates. During the recent Society for Claustrum Research meeting, a panel of experts presented data pertaining to the relationship of these regions and held a discussion on whether the CLA and En should be considered (a) separate unrelated structures, (b) separate nuclei within the same formation, or (c) subregions of a continuous structure. This review article summarizes that discussion, presenting comparisons of the cytoarchitecture, neurochemical profiles, genetic markers, and anatomical connectivity of the CLA and En across several mammalian species. In rodents, we conclude that the CLA and the dorsal endopiriform nucleus (DEn) are subregions of a larger complex, which likely performs analogous computations and exert similar effects on their respective cortical targets (e.g., sensorimotor versus limbic). Moving forward, we recommend that the field retain the nomenclature currently employed for this region but should continue to examine the delineation of these structures across different species. Using thorough descriptions of a variety of anatomical features, this review offers a clear definition of the CLA and En in rodents, which provides a framework for identifying homologous structures in primates.
Collapse
Affiliation(s)
- Jared B. Smith
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kevin D. Alloway
- Neural and Behavioral Sciences, Center for Neural Engineering, Pennsylvania State University, University Park, PA 16802, USA
| | - Patrick R. Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rena Orman
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, 11203 USA
| | - David H. Reser
- Graduate Entry Medicine Program, Monash Rural Health Churchill, Monash University, Churchill, Victoria 3842, Australia
- Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | | | - Glenn D. R. Watson
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| |
Collapse
|
25
|
Kliewer A, Schmiedel F, Sianati S, Bailey A, Bateman JT, Levitt ES, Williams JT, Christie MJ, Schulz S. Phosphorylation-deficient G-protein-biased μ-opioid receptors improve analgesia and diminish tolerance but worsen opioid side effects. Nat Commun 2019; 10:367. [PMID: 30664663 PMCID: PMC6341117 DOI: 10.1038/s41467-018-08162-1] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022] Open
Abstract
Opioid analgesics are powerful pain relievers; however, over time, pain control diminishes as analgesic tolerance develops. The molecular mechanisms initiating tolerance have remained unresolved to date. We have previously shown that desensitization of the μ-opioid receptor and interaction with β-arrestins is controlled by carboxyl-terminal phosphorylation. Here we created knockin mice with a series of serine- and threonine-to-alanine mutations that render the receptor increasingly unable to recruit β-arrestins. Desensitization is inhibited in locus coeruleus neurons of mutant mice. Opioid-induced analgesia is strongly enhanced and analgesic tolerance is greatly diminished. Surprisingly, respiratory depression, constipation, and opioid withdrawal signs are unchanged or exacerbated, indicating that β-arrestin recruitment does not contribute to the severity of opioid side effects and, hence, predicting that G-protein-biased µ-agonists are still likely to elicit severe adverse effects. In conclusion, our findings identify carboxyl-terminal multisite phosphorylation as key step that drives acute μ-opioid receptor desensitization and long-term tolerance.
Collapse
Affiliation(s)
- A Kliewer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747, Jena, Germany
| | - F Schmiedel
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747, Jena, Germany
| | - S Sianati
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, NSW, 2006, Australia
| | - A Bailey
- Institute of Medical and Biomedical Education, St George's University of London, London, SW17 ORE, UK
| | - J T Bateman
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, 32608, USA
| | - E S Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, 32608, USA
| | - J T Williams
- The Vollum Institute, Oregon Health and Science University, 3181S.W. Sam Jackson Pk. Rd., Portland, OR, 97239, USA
| | - M J Christie
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, NSW, 2006, Australia
| | - S Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, 07747, Jena, Germany.
| |
Collapse
|
26
|
Zanos P, Keyworth H, Georgiou P, Hambsch B, Otte DM, Kitchen I, Zimmer A, Bailey A. Chronic nicotine administration restores brain region specific upregulation of oxytocin receptor binding levels in a G72 mouse model of schizophrenia. Eur J Neurosci 2018; 50:2255-2263. [DOI: 10.1111/ejn.14155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/07/2018] [Accepted: 08/26/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Panos Zanos
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
- Department of Psychiatry School of Medicine University of Maryland Baltimore Maryland USA
| | - Helen Keyworth
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
| | - Polymnia Georgiou
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
- Department of Psychiatry School of Medicine University of Maryland Baltimore Maryland USA
| | - Boris Hambsch
- GKM Gesellschaft für Therapieforschung mbH Lessingstraße München Germany
| | - David M. Otte
- Institute for Molecular Psychiatry Medical Faculty University of Bonn Bonn Germany
| | - Ian Kitchen
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
| | - Andreas Zimmer
- Institute for Molecular Psychiatry Medical Faculty University of Bonn Bonn Germany
| | - Alexis Bailey
- Department of Biochemistry and Physiology Faculty of Health and Medical Sciences University of Surrey Guildford Surrey UK
- Institute of Medical and Biomedical Education St George's University of London London SW17 0RE UK
| |
Collapse
|
27
|
Helal MA, Habib ES, Chittiboyina AG. Selective kappa opioid antagonists for treatment of addiction, are we there yet? Eur J Med Chem 2017; 141:632-647. [PMID: 29107424 DOI: 10.1016/j.ejmech.2017.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/06/2017] [Accepted: 10/07/2017] [Indexed: 11/30/2022]
Abstract
Kappa opioid receptor (KOP) is a G-protein coupled receptor mainly expressed in the cerebral cortex and hypothalamus. It is implicated in nociception, diuresis, emotion, cognition, and immune system functions. KOP agonists possess a strong analgesic effect accompanied by a feeling of dysphoria. On the other hand, antagonists of this receptor were found to block depression, anxiety, and drug-seeking behaviors in animal models. Recently, great interest has been given to the development of selective KOP antagonists as an addiction treatment that does not cause dependence itself or show high relapse rates like the currently used agents. This review provides a comprehensive survey of the KOP antagonists developed for this purpose together with their in vivo studies and clinical trials. In addition, a future perspective and recommendations for the work needed to develop clinically relevant KOP antagonists are presented.
Collapse
Affiliation(s)
- Mohamed A Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza 12588, Egypt; Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| | - Eman S Habib
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Amar G Chittiboyina
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| |
Collapse
|
28
|
Opioid-induced rewards, locomotion, and dopamine activation: A proposed model for control by mesopontine and rostromedial tegmental neurons. Neurosci Biobehav Rev 2017; 83:72-82. [PMID: 28951251 DOI: 10.1016/j.neubiorev.2017.09.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/12/2017] [Accepted: 09/21/2017] [Indexed: 01/06/2023]
Abstract
Opioids, such as morphine or heroin, increase forebrain dopamine (DA) release and locomotion, and support the acquisition of conditioned place preference (CPP) or self-administration. The most sensitive sites for these opioid effects in rodents are in the ventral tegmental area (VTA) and rostromedial tegmental nucleus (RMTg). Opioid inhibition of GABA neurons in these sites is hypothesized to lead to arousing and rewarding effects through disinhibition of VTA DA neurons. We review findings that the laterodorsal tegmental (LDTg) and pedunculopontine tegmental (PPTg) nuclei, which each contain cholinergic, GABAergic, and glutamatergic cells, are important for these effects. LDTg and/or PPTg cholinergic inputs to VTA mediate opioid-induced locomotion and DA activation via VTA M5 muscarinic receptors. LDTg and/or PPTg cholinergic inputs to RMTg also modulate opioid-induced locomotion. Lesions or inhibition of LDTg or PPTg neurons reduce morphine-induced increases in forebrain DA release, acquisition of morphine CPP or self-administration. We propose a circuit model that links VTA and RMTg GABA with LDTg and PPTg neurons critical for DA-dependent opioid effects in drug-naïve rodents.
Collapse
|
29
|
Characterization of [ 3 H] oxymorphone binding sites in mouse brain: Quantitative autoradiography in opioid receptor knockout mice. Neurosci Lett 2017; 643:16-21. [DOI: 10.1016/j.neulet.2017.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/01/2017] [Accepted: 02/01/2017] [Indexed: 11/19/2022]
|
30
|
François A, Scherrer G. Delta Opioid Receptor Expression and Function in Primary Afferent Somatosensory Neurons. Handb Exp Pharmacol 2017; 247:87-114. [PMID: 28993838 DOI: 10.1007/164_2017_58] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The functional diversity of primary afferent neurons of the dorsal root ganglia (DRG) generates a variety of qualitatively and quantitatively distinct somatosensory experiences, from shooting pain to pleasant touch. In recent years, the identification of dozens of genetic markers specifically expressed by subpopulations of DRG neurons has dramatically improved our understanding of this diversity and provided the tools to manipulate their activity and uncover their molecular identity and function. Opioid receptors have long been known to be expressed by discrete populations of DRG neurons, in which they regulate cell excitability and neurotransmitter release. We review recent insights into the identity of the DRG neurons that express the delta opioid receptor (DOR) and the ion channel mechanisms that DOR engages in these cells to regulate sensory input. We highlight recent findings derived from DORGFP reporter mice and from in situ hybridization and RNA sequencing studies in wild-type mice that revealed DOR presence in cutaneous mechanosensory afferents eliciting touch and implicated in tactile allodynia. Mechanistically, we describe how DOR modulates opening of voltage-gated calcium channels (VGCCs) to control glutamatergic neurotransmission between somatosensory neurons and postsynaptic neurons in the spinal cord dorsal horn. We additionally discuss other potential signaling mechanisms, including those involving potassium channels, which DOR may engage to fine tune somatosensation. We conclude by discussing how this knowledge may explain the analgesic properties of DOR agonists against mechanical pain and uncovers an unanticipated specialized function for DOR in cutaneous mechanosensation.
Collapse
Affiliation(s)
- Amaury François
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Grégory Scherrer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA. .,Department of Molecular and Cellular Physiology, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA. .,Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
31
|
Moore K, Madularu D, Iriah S, Yee JR, Kulkarni P, Darcq E, Kieffer BL, Ferris CF. BOLD Imaging in Awake Wild-Type and Mu-Opioid Receptor Knock-Out Mice Reveals On-Target Activation Maps in Response to Oxycodone. Front Neurosci 2016; 10:471. [PMID: 27857679 PMCID: PMC5094148 DOI: 10.3389/fnins.2016.00471] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/03/2016] [Indexed: 02/06/2023] Open
Abstract
Blood oxygen level dependent (BOLD) imaging in awake mice was used to identify differences in brain activity between wild-type, and Mu (μ) opioid receptor knock-outs (MuKO) in response to oxycodone (OXY). Using a segmented, annotated MRI mouse atlas and computational analysis, patterns of integrated positive and negative BOLD activity were identified across 122 brain areas. The pattern of positive BOLD showed enhanced activation across the brain in WT mice within 15 min of intraperitoneal administration of 2.5 mg of OXY. BOLD activation was detected in 72 regions out of 122, and was most prominent in areas of high μ opioid receptor density (thalamus, ventral tegmental area, substantia nigra, caudate putamen, basal amygdala, and hypothalamus), and focus on pain circuits indicated strong activation in major pain processing centers (central amygdala, solitary tract, parabrachial area, insular cortex, gigantocellularis area, ventral thalamus primary sensory cortex, and prelimbic cortex). Importantly, the OXY-induced positive BOLD was eliminated in MuKO mice in most regions, with few exceptions (some cerebellar nuclei, CA3 of the hippocampus, medial amygdala, and preoptic areas). This result indicates that most effects of OXY on positive BOLD are mediated by the μ opioid receptor (on-target effects). OXY also caused an increase in negative BOLD in WT mice in few regions (16 out of 122) and, unlike the positive BOLD response the negative BOLD was only partially eliminated in the MuKO mice (cerebellum), and in some case intensified (hippocampus). Negative BOLD analysis therefore shows activation and deactivation events in the absence of the μ receptor for some areas where receptor expression is normally extremely low or absent (off-target effects). Together, our approach permits establishing opioid-induced BOLD activation maps in awake mice. In addition, comparison of WT and MuKO mutant mice reveals both on-target and off-target activation events, and set an OXY brain signature that should, in the future, be compared to other μ opioid agonists.
Collapse
Affiliation(s)
- Kelsey Moore
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University Boston, MA, USA
| | - Dan Madularu
- Brain Imaging Center, Douglas Hospital Research Institute, McGill University Montreal, QC, Canada
| | - Sade Iriah
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University Boston, MA, USA
| | - Jason R Yee
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University Boston, MA, USA
| | - Praveen Kulkarni
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University Boston, MA, USA
| | - Emmanuel Darcq
- Brain Imaging Center, Douglas Hospital Research Institute, McGill University Montreal, QC, Canada
| | - Brigitte L Kieffer
- Brain Imaging Center, Douglas Hospital Research Institute, McGill University Montreal, QC, Canada
| | - Craig F Ferris
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University Boston, MA, USA
| |
Collapse
|
32
|
Pleiotrophin overexpression regulates amphetamine-induced reward and striatal dopaminergic denervation without changing the expression of dopamine D1 and D2 receptors: Implications for neuroinflammation. Eur Neuropsychopharmacol 2016; 26:1794-1805. [PMID: 27642078 DOI: 10.1016/j.euroneuro.2016.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/02/2016] [Accepted: 09/01/2016] [Indexed: 12/22/2022]
Abstract
It was previously shown that mice with genetic deletion of the neurotrophic factor pleiotrophin (PTN-/-) show enhanced amphetamine neurotoxicity and impair extinction of amphetamine conditioned place preference (CPP), suggesting a modulatory role of PTN in amphetamine neurotoxicity and reward. We have now studied the effects of amphetamine (10mg/kg, 4 times, every 2h) in the striatum of mice with transgenic PTN overexpression (PTN-Tg) in the brain and in wild type (WT) mice. Amphetamine caused an enhanced loss of striatal dopaminergic terminals, together with a highly significant aggravation of amphetamine-induced increase in the number of GFAP-positive astrocytes, in the striatum of PTN-Tg mice compared to WT mice. Given the known contribution of D1 and D2 dopamine receptors to the neurotoxic effects of amphetamine, we also performed quantitative receptor autoradiography of both receptors in the brains of PTN-Tg and WT mice. D1 and D2 receptors binding in the striatum and other regions of interest was not altered by genotype or treatment. Finally, we found that amphetamine CPP was significantly reduced in PTN-Tg mice. The data demonstrate that PTN overexpression in the brain blocks the conditioning effects of amphetamine and enhances the characteristic striatal dopaminergic denervation caused by this drug. These results indicate for the first time deleterious effects of PTN in vivo by mechanisms that are probably independent of changes in the expression of D1 and D2 dopamine receptors. The data also suggest that PTN-induced neuroinflammation could be involved in the enhanced neurotoxic effects of amphetamine in the striatum of PTN-Tg mice.
Collapse
|
33
|
Schmitt S, Delamare J, Tirel O, Fillesoye F, Dhilly M, Perrio C. N-[ 18F]-FluoropropylJDTic for κ-opioid receptor PET imaging: Radiosynthesis, pre-clinical evaluation, and metabolic investigation in comparison with parent JDTic. Nucl Med Biol 2016; 44:50-61. [PMID: 27821345 DOI: 10.1016/j.nucmedbio.2016.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/07/2016] [Accepted: 09/17/2016] [Indexed: 02/08/2023]
Abstract
INTRODUCTION To image kappa opioid receptor (KOR) for preclinical studies, N-fluoropropylJDTic 9 derived from the best-established KOR antagonist JDTic, was labeled with fluorine-18. METHODS Radiosynthesis of [18F]9 was achieved according to an automated two-step procedure from [18F]-fluoride. Peripheral and cerebral distributions were determined by ex vivo experiments and by PET imaging in mouse. Radiometabolism studies were performed both in vivo in mice and in vitro in mouse and human liver microsomes. Identification of the major metabolic fragmentations was carried out by UPLC-MS analysis of enzymatic cleavage of non-radioactive ligand 9. Microsomal metabolic degradation of parent JDTic was also achieved for comparison. RESULTS The radiotracer [18F]9 was produced after 140±5min total synthesis time (2.2±0.4% not decay corrected radiochemical yield) with a specific activity of 41-89GBq/μmol (1.1-2.4Ci/μmol). Peripheral and regional brain distributions of [18F]9 were consistent with known KOR locations but no significant specific binding in brain was shown. [18F]9 presented a typical hepatobiliary and renal elimination, and was rapidly metabolized. The in vivo and in vitro radiometabolic profiles of [18F]9 were similar. Piperidine 12 was identified as the major metabolic fragment of the non-radioactive ligand 9. JDTic 7 was found to be much more stable than 9. CONCLUSION Although the newly proposed radioligand [18F]9 was concluded to be not suitable for KOR PET imaging due to the formation of brain penetrating radiometabolites, our findings highlight the metabolic stability of JDTic and may help in the design of novel JDTic derivatives for in vivo applications.
Collapse
Affiliation(s)
- Sébastien Schmitt
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Jérôme Delamare
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Olivier Tirel
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Fabien Fillesoye
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Martine Dhilly
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Cécile Perrio
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France.
| |
Collapse
|
34
|
Thanos PK, Zhuo J, Robison L, Kim R, Ananth M, Choai I, Grunseich A, Grissom NM, George R, Delis F, Reyes TM. Suboptimal maternal diets alter mu opioid receptor and dopamine type 1 receptor binding but exert no effect on dopamine transporters in the offspring brain. Int J Dev Neurosci 2016; 64:21-28. [PMID: 27666382 DOI: 10.1016/j.ijdevneu.2016.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/07/2016] [Accepted: 09/15/2016] [Indexed: 11/18/2022] Open
Abstract
Birthweight is a marker for suboptimal fetal growth and development in utero. Offspring can be born large for gestational age (LGA), which is linked to maternal obesity or excessive gestational weight gain, as well as small for gestational age (SGA), arising from nutrient or calorie deficiency, placental dysfunction, or other maternal conditions (hypertension, infection). In humans, LGA and SGA babies are at an increased risk for certain neurodevelopmental disorders, including Attention Deficit/Hyperactivity Disorder, schizophrenia, and social and mood disorders. Using mouse models of LGA (maternal high fat (HF) diet) and SGA (maternal low protein (LP) diet) offspring, our lab has previously shown that these offspring display alterations in the expression of mesocorticolimbic genes that regulate dopamine and opioid function, thus indicating that these brain regions and neurotransmitter systems are vulnerable to gestational insults. Interestingly, these two maternal diets affected dopamine and opioid systems in somewhat opposing directions (e.g., LP offspring are generally hyperdopaminergic with reduced opioid expression, and the reverse is found for the HF offspring). These data largely involved evaluation at the transcriptional level, so the present experiment was designed to extend these analyses through an assessment of receptor binding. In this study, control, SGA and LGA offspring were generated from dams fed control, low protein or high fat diet, respectively, throughout pregnancy and lactation. At weaning, mice were placed on the control diet and sacrificed at 12 weeks of age. In vitro autoradiography was used to measure mu-opioid receptor (MOR), dopamine type 1 receptor (D1R), and dopamine transporter (DAT) binding level in mesolimbic brain regions. Results showed that the LP offspring (males and females) had significantly higher MOR and D1R binding than the control animals in the regions associated with reward. In HF offspring there were no differences in MOR binding, and limited increases in D1R binding, seen only in females in the nucleus accumbens core and the dorsomedial caudate/putamen. DAT binding revealed no differences in either models. In conclusion, LP but not HF offspring show significantly elevated MOR and D1R binding in the brain thus affecting DA and opioid signaling. These findings advance the current understanding of how suboptimal gestational diets can adversely impact neurodevelopment and increase the risk for disorders such as ADHD, obesity and addiction.
Collapse
Affiliation(s)
- Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Research Institute on Addictions, University at Buffalo, 14203, USA.
| | - Jianmin Zhuo
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Research Institute on Addictions, University at Buffalo, 14203, USA
| | - Lisa Robison
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Research Institute on Addictions, University at Buffalo, 14203, USA
| | - Ronald Kim
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Research Institute on Addictions, University at Buffalo, 14203, USA
| | - Mala Ananth
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Research Institute on Addictions, University at Buffalo, 14203, USA
| | - Ilon Choai
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Research Institute on Addictions, University at Buffalo, 14203, USA
| | - Adam Grunseich
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Research Institute on Addictions, University at Buffalo, 14203, USA
| | - Nicola M Grissom
- Department of Psychiatry and Behavioral Neurosciences, University of Cincinnati, Cincinnati, OH 45237, USA
| | - Robert George
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Foteini Delis
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Research Institute on Addictions, University at Buffalo, 14203, USA
| | - Teresa M Reyes
- Department of Psychiatry and Behavioral Neurosciences, University of Cincinnati, Cincinnati, OH 45237, USA
| |
Collapse
|
35
|
Wright SR, Zanos P, Georgiou P, Yoo JH, Ledent C, Hourani SM, Kitchen I, Winsky-Sommerer R, Bailey A. A critical role of striatal A2A R-mGlu5 R interactions in modulating the psychomotor and drug-seeking effects of methamphetamine. Addict Biol 2016; 21:811-25. [PMID: 25975203 DOI: 10.1111/adb.12259] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Addiction to psychostimulants is a major public health problem with no available treatment. Adenosine A2A receptors (A2A R) co-localize with metabotropic glutamate 5 receptors (mGlu5 R) in the striatum and functionally interact to modulate behaviours induced by addictive substances, such as alcohol. Using genetic and pharmacological antagonism of A2A R in mice, we investigated whether A2A R-mGlu5 R interaction can regulate the locomotor, stereotypic and drug-seeking effect of methamphetamine and cocaine, two drugs that exhibit distinct mechanism of action. Genetic deletion of A2A R, as well as combined administration of sub-threshold doses of the selective A2A R antagonist (SCH 58261, 0.01 mg/kg, i.p.) with the mGlu5 R antagonist, 3-((2-methyl-4-thiazolyl)ethynyl)pyridine (0.01 mg/kg, i.p.), prevented methamphetamine- but not cocaine-induced hyperactivity and stereotypic rearing behaviour. This drug combination also prevented methamphetamine-rewarding effects in a conditioned-place preference paradigm. Moreover, mGlu5 R binding was reduced in the nucleus accumbens core of A2A R knockout (KO) mice supporting an interaction between these receptors in a brain region crucial in mediating addiction processes. Chronic methamphetamine, but not cocaine administration, resulted in a significant increase in striatal mGlu5 R binding in wild-type mice, which was absent in the A2A R KO mice. These data are in support of a critical role of striatal A2A R-mGlu5 R functional interaction in mediating the ambulatory, stereotypic and reinforcing effects of methamphetamine but not cocaine-induced hyperlocomotion or stereotypy. The present study highlights a distinct and selective mechanistic role for this receptor interaction in regulating methamphetamine-induced behaviours and suggests that combined antagonism of A2A R and mGlu5 R may represent a novel therapy for methamphetamine addiction.
Collapse
Affiliation(s)
- Sherie R. Wright
- Sleep, Chronobiology and Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Panos Zanos
- Sleep, Chronobiology and Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Polymnia Georgiou
- Sleep, Chronobiology and Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Ji-Hoon Yoo
- Sleep, Chronobiology and Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Catherine Ledent
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire; Université Libre de Bruxelles; Belgium
| | - Susanna M. Hourani
- Sleep, Chronobiology and Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Ian Kitchen
- Sleep, Chronobiology and Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Raphaelle Winsky-Sommerer
- Sleep, Chronobiology and Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Alexis Bailey
- Sleep, Chronobiology and Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| |
Collapse
|
36
|
Watson GDR, Smith JB, Alloway KD. Interhemispheric connections between the infralimbic and entorhinal cortices: The endopiriform nucleus has limbic connections that parallel the sensory and motor connections of the claustrum. J Comp Neurol 2016; 525:1363-1380. [PMID: 26860547 DOI: 10.1002/cne.23981] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/17/2022]
Abstract
We have previously shown that the claustrum is part of an interhemispheric circuit that interconnects somesthetic-motor and visual-motor cortical regions. The role of the claustrum in processing limbic information, however, is poorly understood. Some evidence suggests that the dorsal endopiriform nucleus (DEn), which lies immediately ventral to the claustrum, has connections with limbic cortical areas and should be considered part of a claustrum-DEn complex. To determine whether DEn has similar patterns of cortical connections as the claustrum, we used anterograde and retrograde tracing techniques to elucidate the connectivity of DEn. Following injections of retrograde tracers into DEn, labeled neurons appeared bilaterally in the infralimbic (IL) cortex and ipsilaterally in the entorhinal and piriform cortices. Anterograde tracer injections in DEn revealed labeled terminals in the same cortical regions, but only in the ipsilateral hemisphere. These tracer injections also revealed extensive longitudinal projections throughout the rostrocaudal extent of the nucleus. Dual retrograde tracer injections into IL and lateral entorhinal cortex (LEnt) revealed intermingling of labeled neurons in ipsilateral DEn, including many double-labeled neurons. In other experiments, anterograde and retrograde tracers were separately injected into IL of each hemisphere of the same animal. This revealed an interhemispheric circuit in which IL projects bilaterally to DEn, with the densest terminal labeling appearing in the contralateral hemisphere around retrogradely labeled neurons that project to IL in that hemisphere. By showing that DEn and claustrum have parallel sets of connections, these results suggest that DEn and claustrum perform similar functions in processing limbic and sensorimotor information, respectively. J. Comp. Neurol. 525:1363-1380, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Glenn D R Watson
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033.,Center for Neural Engineering, Pennsylvania State University, University Park, PA 16802
| | - Jared B Smith
- Center for Neural Engineering, Pennsylvania State University, University Park, PA 16802.,Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA 16802
| | - Kevin D Alloway
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033.,Center for Neural Engineering, Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
37
|
Maternal Weaning Modulates Emotional Behavior and Regulates the Gut-Brain Axis. Sci Rep 2016; 6:21958. [PMID: 26903212 PMCID: PMC4763306 DOI: 10.1038/srep21958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/02/2016] [Indexed: 12/19/2022] Open
Abstract
Evidence shows that nutritional and environmental stress stimuli during postnatal period influence brain development and interactions between gut and brain. In this study we show that in rats, prevention of weaning from maternal milk results in depressive-like behavior, which is accompanied by changes in the gut bacteria and host metabolism. Depressive-like behavior was studied using the forced-swim test on postnatal day (PND) 25 in rats either weaned on PND 21, or left with their mother until PND 25 (non-weaned). Non-weaned rats showed an increased immobility time consistent with a depressive phenotype. Fluorescence in situ hybridization showed non-weaned rats to harbor significantly lowered Clostridium histolyticum bacterial groups but exhibit marked stress-induced increases. Metabonomic analysis of urine from these animals revealed significant differences in the metabolic profiles, with biochemical phenotypes indicative of depression in the non-weaned animals. In addition, non-weaned rats showed resistance to stress-induced modulation of oxytocin receptors in amygdala nuclei, which is indicative of passive stress-coping mechanism. We conclude that delaying weaning results in alterations to the gut microbiota and global metabolic profiles which may contribute to a depressive phenotype and raise the issue that mood disorders at early developmental ages may reflect interplay between mammalian host and resident bacteria.
Collapse
|
38
|
Zanos P, Georgiou P, Gonzalez LR, Hourani S, Chen Y, Kitchen I, Kieffer BL, Winsky-Sommerer R, Bailey A. Emotional Impairment and Persistent Upregulation of mGlu5 Receptor following Morphine Abstinence: Implications of an mGlu5-MOPr Interaction. Int J Neuropsychopharmacol 2016; 19:pyw011. [PMID: 26861145 PMCID: PMC4966274 DOI: 10.1093/ijnp/pyw011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 02/02/2016] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND A difficult problem in treating opioid addicts is the maintenance of a drug-free state because of the negative emotional symptoms associated with withdrawal, which may trigger relapse. Several lines of evidence suggest a role for the metabotropic glutamate receptor 5 in opioid addiction; however, its involvement during opioid withdrawal is not clear. METHODS Mice were treated with a 7-day escalating-dose morphine administration paradigm. Following withdrawal, the development of affective behaviors was assessed using the 3-chambered box, open-field, elevated plus-maze and forced-swim tests. Metabotropic glutamate receptor 5 autoradiographic binding was performed in mouse brains undergoing chronic morphine treatment and 7 days withdrawal. Moreover, since there is evidence showing direct effects of opioid drugs on the metabotropic glutamate receptor 5 system, the presence of an metabotropic glutamate receptor 5/μ-opioid receptor interaction was assessed by performing metabotropic glutamate receptor 5 autoradiographic binding in brains of mice lacking the μ-opioid receptor gene. RESULTS Withdrawal from chronic morphine administration induced anxiety-like, depressive-like, and impaired sociability behaviors concomitant with a marked upregulation of metabotropic glutamate receptor 5 binding. Administration of the metabotropic glutamate receptor 5 antagonist, 3-((2-Methyl-4-thiazolyl)ethynyl)pyridine, reversed morphine abstinence-induced depressive-like behaviors. A brain region-specific increase in metabotropic glutamate receptor 5 binding was observed in the nucleus accumbens shell, thalamus, hypothalamus, and amygdala of μ-opioid receptor knockout mice compared with controls. CONCLUSIONS These results suggest an association between metabotropic glutamate receptor 5 alterations and the emergence of opioid withdrawal-related affective behaviors. This study supports metabotropic glutamate receptor 5 system as a target for the development of pharmacotherapies for the treatment of opioid addiction. Moreover, our data show direct effects of μ-opioid receptor system manipulation on metabotropic glutamate receptor 5 binding in the brain.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Alexis Bailey
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK (Dr Zanos, Dr Georgiou, Ms Rojo Gonzalez, Prof. Hourani, Prof. Kitchen, Dr Winsky-Sommerer, and Dr Bailey); Department of Psychiatry, University of Maryland, School of Medicine, Baltimore, MD (Dr Zanos and Dr Georgiou); Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK (Dr Chen); Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Illkirch, France (Prof. Kieffer); Douglas Hospital Research Center, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada (Prof. Kieffer); Institute of Medical and Biomedical Education, St George's University of London, London (Dr Bailey).
| |
Collapse
|
39
|
Pellissier LP, Pujol CN, Becker JAJ, Le Merrer J. Delta Opioid Receptors: Learning and Motivation. Handb Exp Pharmacol 2016; 247:227-260. [PMID: 28035528 DOI: 10.1007/164_2016_89] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Delta opioid receptor (DOR) displays a unique, highly conserved, structure and an original pattern of distribution in the central nervous system, pointing to a distinct and specific functional role among opioid peptide receptors. Over the last 15 years, in vivo pharmacology and genetic models have allowed significant advances in the understanding of this role. In this review, we will focus on the involvement of DOR in modulating different types of hippocampal- and striatal-dependent learning processes as well as motor function, motivation, and reward. Remarkably, DOR seems to play a key role in balancing hippocampal and striatal functions, with major implications for the control of cognitive performance and motor function under healthy and pathological conditions.
Collapse
Affiliation(s)
- L P Pellissier
- Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, INSERM, Université François Rabelais, IFCE, 37380, Nouzilly, France
| | - C N Pujol
- Département de Neurosciences, Institut de Génomique fonctionnelle, INSERM U-661, CNRS UMR-5203, 34094, Montpellier, France
| | - J A J Becker
- Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, INSERM, Université François Rabelais, IFCE, 37380, Nouzilly, France
| | - J Le Merrer
- Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, INSERM, Université François Rabelais, IFCE, 37380, Nouzilly, France.
| |
Collapse
|
40
|
Peppin JF, Raffa RB. Delta opioid agonists: a concise update on potential therapeutic applications. J Clin Pharm Ther 2015; 40:155-66. [PMID: 25726896 DOI: 10.1111/jcpt.12244] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/24/2014] [Indexed: 01/23/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE The endogenous opioid system co-evolved with chemical defences, or at times symbiotic relationships, between plants and other autotrophs and heterotrophic predators - thus, it is not surprising that endogenous opioid ligands and exogenous mimetic ligands produce diverse physiological effects. Among the endogenous opioid peptides (endomorphins, enkephalins, dynorphins and nociception/orphanin FQ) derived from the precursors encoded by four genes (PNOC, PENK, PDYN and POMC) are the pentapeptides Met-enkephalin (Tyr-Gly-Gly-Phe-Met) and Leu-enkephalin (Tyr-Gly-Gly-Phe-Leu). The physiological effects of the enkephalins are mediated via 7-transmembrane G protein-coupled receptors, including delta opioid receptor (DOR). We present a concise update on the status of progress and opportunities of this approach. METHODS A literature search of the PUBMED database and a combination of keywords including delta opioid receptor, analgesia, mood and individual compounds identified therein, from industry and other source, and from www.clinicaltrials.com. RESULTS AND DISCUSSION DOR agonist and antagonist ligands have been developed with ever increasing affinity and selectivity for DOR over other opioid receptor subtypes and studied for therapeutic utility, primarily for pain relief, but also for other clinical endpoints. WHAT IS NEW AND CONCLUSION Selective DOR agonists have been designed with a large increase in therapeutic window for a variety of potential CNS applications including pain, depression, and learning and memory among others.
Collapse
Affiliation(s)
- J F Peppin
- Center for Bioethics, Pain Management and Medicine, University City, MO, USA; Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | | |
Collapse
|
41
|
Abstract
UNLABELLED The nociceptin/orphanin FQ (NOP) receptor, the fourth member of the opioid receptor family, is involved in many processes common to the opioid receptors including pain and drug abuse. To better characterize receptor location and trafficking, knock-in mice were created by inserting the gene encoding enhanced green fluorescent protein (eGFP) into the NOP receptor gene (Oprl1) and producing mice expressing a functional NOP-eGFP C-terminal fusion in place of the native NOP receptor. The NOP-eGFP receptor was present in brain of homozygous knock-in animals in concentrations somewhat higher than in wild-type mice and was functional when tested for stimulation of [(35)S]GTPγS binding in vitro and in patch-clamp electrophysiology in dorsal root ganglia (DRG) neurons and hippocampal slices. Inhibition of morphine analgesia was equivalent when tested in knock-in and wild-type mice. Imaging revealed detailed neuroanatomy in brain, spinal cord, and DRG and was generally consistent with in vitro autoradiographic imaging of receptor location. Multicolor immunohistochemistry identified cells coexpressing various spinal cord and DRG cellular markers, as well as coexpression with μ-opioid receptors in DRG and brain regions. Both in tissue slices and primary cultures, the NOP-eGFP receptors appear throughout the cell body and in processes. These knock-in mice have NOP receptors that function both in vitro and in vivo and appear to be an exceptional tool to study receptor neuroanatomy and correlate with NOP receptor function. SIGNIFICANCE STATEMENT The NOP receptor, the fourth member of the opioid receptor family, is involved in pain, drug abuse, and a number of other CNS processes. The regional and cellular distribution has been difficult to determine due to lack of validated antibodies for immunohistochemical analysis. To provide a new tool for the investigation of receptor localization, we have produced knock-in mice with a fluorescent-tagged NOP receptor in place of the native NOP receptor. These knock-in mice have NOP receptors that function both in vitro and in vivo and have provided a detailed characterization of NOP receptors in brain, spinal cord, and DRG neurons. They appear to be an exceptional tool to study receptor neuroanatomy and correlate with NOP receptor function.
Collapse
|
42
|
Georgiou P, Zanos P, Ehteramyan M, Hourani S, Kitchen I, Maldonado R, Bailey A. Differential regulation of mGlu5 R and ΜOPr by priming- and cue-induced reinstatement of cocaine-seeking behaviour in mice. Addict Biol 2015; 20:902-12. [PMID: 25522112 DOI: 10.1111/adb.12208] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The key problem for the treatment of drug addiction is relapse to drug use after abstinence that can be triggered by drug-associated cues, re-exposure to the drug itself and stress. Understanding the neurobiological mechanisms underlying relapse is essential in order to develop effective pharmacotherapies for its prevention. Given the evidence implicating the metabotropic glutamate receptor 5 (mGlu5 R), μ-opioid receptor (MOPr), κ-opioid receptor (ΚOPr) and oxytocin receptor (OTR) systems in cocaine addiction and relapse, our aim was to assess the modulation of these receptors using a mouse model of cue- and priming-induced reinstatement of cocaine seeking. Male mice were trained to self-administer cocaine (1 mg/kg/infusion, i.v.) and were randomized into different groups: (1) cocaine self-administration; (2) cocaine extinction; (3) cocaine-primed (10 mg/kg i.p.); or (4) cue-induced reinstatement of cocaine seeking. Mice undergoing the same protocols but receiving saline instead of cocaine were used as controls. Quantitative autoradiography of mGlu5 R, MOPr, KOPr and OTR showed a persistent cocaine-induced upregulation of the mGlu5 R and OTR in the lateral septum and central amygdala, respectively. Moreover, a downregulation of mGlu5 R and MOPr was observed in the basolateral amygdala and striatum, respectively. Further, we showed that priming- but not cue-induced reinstatement upregulates mGlu5 R and MOPr binding in the nucleus accumbens core and basolateral amygdala, respectively, while cue- but not priming-induced reinstatement downregulates MOPr binding in caudate putamen and nucleus accumbens core. This is the first study to provide direct evidence of reinstatement-induced receptor alterations that are likely to contribute to the neurobiological mechanisms underpinning relapse to cocaine seeking.
Collapse
Affiliation(s)
- Polymnia Georgiou
- Sleep, Chronobiology & Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Panos Zanos
- Sleep, Chronobiology & Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Mazdak Ehteramyan
- Department of Experimental and Health Sciences; University of Pompeu Fabra; Spain
| | - Susanna Hourani
- Sleep, Chronobiology & Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Ian Kitchen
- Sleep, Chronobiology & Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| | - Rafael Maldonado
- Department of Experimental and Health Sciences; University of Pompeu Fabra; Spain
| | - Alexis Bailey
- Sleep, Chronobiology & Addiction Group; School of Biosciences and Medicine; Faculty of Health and Medical Sciences; University of Surrey; UK
| |
Collapse
|
43
|
Almatroudi A, Husbands SM, Bailey CP, Bailey SJ. Combined administration of buprenorphine and naltrexone produces antidepressant-like effects in mice. J Psychopharmacol 2015; 29:812-21. [PMID: 26045511 PMCID: PMC5075030 DOI: 10.1177/0269881115586937] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Opiates have been used historically for the treatment of depression. Renewed interest in the use of opiates as antidepressants has focused on the development of kappa opioid receptor (κ-receptor) antagonists. Buprenorphine acts as a partial µ-opioid receptor agonist and a κ-receptor antagonist. By combining buprenorphine with the opioid antagonist naltrexone, the activation of µ-opioid receptors will be reduced and the κ-antagonist properties enhanced. We have established that a combination dose of buprenorphine (1 mg/kg) with naltrexone (1 mg/kg) functions as a short-acting κ-antagonist in the mouse tail withdrawal test. Furthermore, this dose combination is neither rewarding nor aversive in the conditioned place preference paradigm, and is without significant locomotor effects. We have shown for the first time that systemic co-administration of buprenorphine (1 mg/kg) with naltrexone (1 mg/kg) in CD-1 mice produced an antidepressant-like response in behaviours in both the forced swim test and novelty induced hypophagia task. Behaviours in the elevated plus maze and light dark box were not significantly altered by treatment with buprenorphine alone, or in combination with naltrexone. We propose that the combination of buprenorphine with naltrexone represents a novel, and potentially a readily translatable approach, to the treatment of depression.
Collapse
Affiliation(s)
| | | | | | - Sarah J Bailey
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| |
Collapse
|
44
|
A novel anxiogenic role for the delta opioid receptor expressed in GABAergic forebrain neurons. Biol Psychiatry 2015; 77:404-15. [PMID: 25444168 PMCID: PMC4297504 DOI: 10.1016/j.biopsych.2014.07.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 07/15/2014] [Accepted: 07/24/2014] [Indexed: 01/10/2023]
Abstract
BACKGROUND The delta opioid receptor (DOR) is broadly expressed throughout the nervous system; it regulates chronic pain, emotional responses, motivation, and memory. Neural circuits underlying DOR activities have been poorly explored by genetic approaches. We used conditional mouse mutagenesis to elucidate receptor function in GABAergic neurons of the forebrain. METHODS We characterized DOR distribution in the brain of Dlx5/6-CreXOprd1(fl/fl) (Dlx-DOR) mice and tested main central DOR functions through behavioral testing. RESULTS The DOR proteins were strongly deleted in olfactory bulb and striatum and remained intact in cortex and basolateral amygdala. Olfactory perception, circadian activity, and despair-like behaviors were unchanged. In contrast, locomotor stimulant effects of SNC80 (DOR agonist) and SKF81297 (D1 agonist) were abolished and increased, respectively. The Dlx-DOR mice showed lower levels of anxiety in the elevated plus maze, opposing the known high anxiety in constitutive DOR knockout animals. Also, Dlx-DOR mice reached the food more rapidly in a novelty suppressed feeding task, despite their lower motivation for food reward observed in an operant paradigm. Finally, c-fos protein staining after novelty suppressed feeding was strongly reduced in amygdala, concordant with the low anxiety phenotype of Dlx-DOR mice. CONCLUSIONS We demonstrate that DORs expressed in the forebrain mediate the described locomotor effect of SNC80 and inhibit D1-stimulated hyperactivity. Our data also reveal an unanticipated anxiogenic role for this particular DOR subpopulation, with a potential novel adaptive role. In emotional responses, DORs exert dual anxiolytic and anxiogenic roles, both of which may have implications in the area of anxiety disorders.
Collapse
|
45
|
Befort K. Interactions of the opioid and cannabinoid systems in reward: Insights from knockout studies. Front Pharmacol 2015; 6:6. [PMID: 25698968 PMCID: PMC4318341 DOI: 10.3389/fphar.2015.00006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/08/2015] [Indexed: 12/14/2022] Open
Abstract
The opioid system consists of three receptors, mu, delta, and kappa, which are activated by endogenous opioid peptides (enkephalins, endorphins, and dynorphins). The endogenous cannabinoid system comprises lipid neuromodulators (endocannabinoids), enzymes for their synthesis and their degradation and two well-characterized receptors, cannabinoid receptors CB1 and CB2. These systems play a major role in the control of pain as well as in mood regulation, reward processing and the development of addiction. Both opioid and cannabinoid receptors are coupled to G proteins and are expressed throughout the brain reinforcement circuitry. Extending classical pharmacology, research using genetically modified mice has provided important progress in the identification of the specific contribution of each component of these endogenous systems in vivo on reward process. This review will summarize available genetic tools and our present knowledge on the consequences of gene knockout on reinforced behaviors in both systems, with a focus on their potential interactions. A better understanding of opioid-cannabinoid interactions may provide novel strategies for therapies in addicted individuals.
Collapse
Affiliation(s)
- Katia Befort
- CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives - UMR7364, Faculté de Psychologie, Neuropôle de Strasbourg - Université de Strasbourg, Strasbourg France
| |
Collapse
|
46
|
Ceredig RA, Massotte D. Fluorescent knock-in mice to decipher the physiopathological role of G protein-coupled receptors. Front Pharmacol 2015; 5:289. [PMID: 25610398 PMCID: PMC4284998 DOI: 10.3389/fphar.2014.00289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/12/2014] [Indexed: 12/21/2022] Open
Abstract
G protein-coupled receptors (GPCRs) modulate most physiological functions but are also critically involved in numerous pathological states. Approximately a third of marketed drugs target GPCRs, which places this family of receptors in the main arena of pharmacological pre-clinical and clinical research. The complexity of GPCR function demands comprehensive appraisal in native environment to collect in-depth knowledge of receptor physiopathological roles and assess the potential of therapeutic molecules. Identifying neurons expressing endogenous GPCRs is therefore essential to locate them within functional circuits whereas GPCR visualization with subcellular resolution is required to get insight into agonist-induced trafficking. Both remain frequently poorly investigated because direct visualization of endogenous receptors is often hampered by the lack of appropriate tools. Also, monitoring intracellular trafficking requires real-time visualization to gather in-depth knowledge. In this context, knock-in mice expressing a fluorescent protein or a fluorescent version of a GPCR under the control of the endogenous promoter not only help to decipher neuroanatomical circuits but also enable real-time monitoring with subcellular resolution thus providing invaluable information on their trafficking in response to a physiological or a pharmacological challenge. This review will present the animal models and discuss their contribution to the understanding of the physiopathological role of GPCRs. We will also address the drawbacks associated with this methodological approach and browse future directions.
Collapse
Affiliation(s)
- Rhian A Ceredig
- CNRS, Institut des Neurosciences Cellulaires et Intégratives, UPR 3212 Strasbourg, France
| | - Dominique Massotte
- CNRS, Institut des Neurosciences Cellulaires et Intégratives, UPR 3212 Strasbourg, France
| |
Collapse
|
47
|
Nubukpo P. [Place of the opioid system in biology and treatment of Alcohol Use Disorder]. Encephale 2014; 40:457-67. [PMID: 25454364 DOI: 10.1016/j.encep.2014.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 10/03/2014] [Indexed: 11/28/2022]
Abstract
While the DSM 5 has formalized the terminology "Alcohol Use Disorders" (AUD) or "disorders of the use of alcohol" (UAW French translation in progress), the term "alcohol dependence" still used in ICD-10, apriority in the future ICD-11 and above in clinical practice. Addiction to alcohol is the cause of mortality and major morbidity. In terms of therapeutic strategies for its management, alongside the maintenance of abstinence after withdrawal (with a high rate of relapse), the reduction of alcohol consumption below certain thresholds of intake is emerging in order to reduce risk, improve health and regain control of consumption even be an intermediate step towards abstinence. The role of the endogenous opioid system in the modulation of the activity of dopaminergic neurons from the circuit of reward and motivation is well established. An unsteadiness of this system has been described in the alcohol dependence. Indeed, a hypofunction of the endorphin pathway and its mu receptor and a hyperactivity of the dynorphin pathway and its kappa receptor participate in the alcohol reinforcing effects (especially positive and negative). The development of active molecules in this system allows better management of alcohol dependence. Besides naltrexone (mu antagonist) allowed in the maintenance of abstinence after withdrawal, another molecule (nalmefene) with modulating properties of μ and κ opioid receptors is the first drug having obtained an MA in reducing consumption in adult patients with alcohol dependence. Its modulating original pharmacological properties by targeting both the positive but also the negative reinforcing effects of alcohol, are responsible for its development in reducing consumption in the alcohol dependence.
Collapse
Affiliation(s)
- P Nubukpo
- Pôle d'addictologie en Limousin, centre hospitalier Esquirol, 15, rue du Dr-Marcland, 87025 Limoges, France; Pôle de psychiatrie adulte 23G01, centre hospitalier La Valette, 23320 Saint-Vaury, France; UMR/Inserm 1094 NET, faculté de médecine, CHU de Limoges, 2, rue du Dr-Marcland, 87025 Limoges, France.
| |
Collapse
|
48
|
Cahill CM, Taylor AMW, Cook C, Ong E, Morón JA, Evans CJ. Does the kappa opioid receptor system contribute to pain aversion? Front Pharmacol 2014; 5:253. [PMID: 25452729 PMCID: PMC4233910 DOI: 10.3389/fphar.2014.00253] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 10/30/2014] [Indexed: 01/18/2023] Open
Abstract
The kappa opioid receptor (KOR) and the endogenous peptide-ligand dynorphin have received significant attention due the involvement in mediating a variety of behavioral and neurophysiological responses, including opposing the rewarding properties of drugs of abuse including opioids. Accumulating evidence indicates this system is involved in regulating states of motivation and emotion. Acute activation of the KOR produces an increase in motivational behavior to escape a threat, however, KOR activation associated with chronic stress leads to the expression of symptoms indicative of mood disorders. It is well accepted that KOR can produce analgesia and is engaged in chronic pain states including neuropathic pain. Spinal studies have revealed KOR-induced analgesia in reversing pain hypersensitivities associated with peripheral nerve injury. While systemic administration of KOR agonists attenuates nociceptive sensory transmission, this effect appears to be a stress-induced effect as anxiolytic agents, including delta opioid receptor agonists, mitigate KOR agonist-induced analgesia. Additionally, while the role of KOR and dynorphin in driving the dysphoric and aversive components of stress and drug withdrawal has been well characterized, how this system mediates the negative emotional states associated with chronic pain is relatively unexplored. This review provides evidence that dynorphin and the KOR system contribute to the negative affective component of pain and that this receptor system likely contributes to the high comorbidity of mood disorders associated with chronic neuropathic pain.
Collapse
Affiliation(s)
- Catherine M Cahill
- Department of Anesthesiology and Perioperative Care, University of California Irvine Irvine, CA, USA ; Department of Pharmacology, University of California Irvine Irvine, CA, USA ; Department of Biomedical and Molecular Sciences, Queen's University Kingston, ON, Canada
| | - Anna M W Taylor
- Department of Anesthesiology and Perioperative Care, University of California Irvine Irvine, CA, USA ; Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles Los Angeles, CA, USA
| | - Christopher Cook
- Department of Anesthesiology and Perioperative Care, University of California Irvine Irvine, CA, USA ; Department of Pharmacology, University of California Irvine Irvine, CA, USA
| | - Edmund Ong
- Department of Anesthesiology and Perioperative Care, University of California Irvine Irvine, CA, USA ; Department of Biomedical and Molecular Sciences, Queen's University Kingston, ON, Canada
| | - Jose A Morón
- Department of Anesthesiology, Columbia University Medical Center, New York, NY USA
| | - Christopher J Evans
- Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles Los Angeles, CA, USA
| |
Collapse
|
49
|
Gardon O, Faget L, Chu Sin Chung P, Matifas A, Massotte D, Kieffer BL. Expression of mu opioid receptor in dorsal diencephalic conduction system: new insights for the medial habenula. Neuroscience 2014; 277:595-609. [PMID: 25086313 PMCID: PMC4164589 DOI: 10.1016/j.neuroscience.2014.07.053] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/21/2014] [Accepted: 07/21/2014] [Indexed: 11/29/2022]
Abstract
The habenular complex, encompassing medial (MHb) and lateral (LHb) divisions, is a highly conserved epithalamic structure involved in the dorsal diencephalic conduction system (DDC). These brain nuclei regulate information flow between the limbic forebrain and the mid- and hindbrain, integrating cognitive with emotional and sensory processes. The MHb is also one of the strongest expression sites for mu opioid receptors (MORs), which mediate analgesic and rewarding properties of opiates. At present however, anatomical distribution and function of these receptors have been poorly studied in MHb pathways. Here we took advantage of a newly generated MOR-mcherry knock-in mouse line to characterize MOR expression sites in the DDC. MOR-mcherry fluorescent signal is weak in the LHb, but strong expression is visible in the MHb, fasciculus retroflexus (fr) and interpeduncular nucleus (IPN), indicating that MOR is mainly present in the MHb-IPN pathway. MOR-mcherry cell bodies are detected both in basolateral and apical parts of MHb, where the receptor co-localizes with cholinergic and substance P (SP) neurons, respectively, representing two main MHb neuronal populations. MOR-mcherry is expressed in most MHb-SP neurons, and is present in only a subpopulation of MHb-cholinergic neurons. Intense diffuse fluorescence detected in lateral and rostral parts of the IPN further suggests that MOR-mcherry is transported to terminals of these SP and cholinergic neurons. Finally, MOR-mcherry is present in septal regions projecting to the MHb, and in neurons of the central and intermediate IPN. Together, this study describes MOR expression in several compartments of the MHb-IPN circuitry. The remarkably high MOR density in the MHb-IPN pathway suggests that these receptors are in a unique position to mediate analgesic, autonomic and reward responses.
Collapse
Affiliation(s)
- O Gardon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, F-67404 Illkirch, France
| | - L Faget
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, F-67404 Illkirch, France
| | - P Chu Sin Chung
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, F-67404 Illkirch, France
| | - A Matifas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, F-67404 Illkirch, France
| | - D Massotte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, F-67404 Illkirch, France
| | - B L Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/Université de Strasbourg, 1 rue Laurent Fries, F-67404 Illkirch, France.
| |
Collapse
|
50
|
Sasaki K, Sumiyoshi A, Nonaka H, Kasahara Y, Ikeda K, Hall FS, Uhl GR, Watanabe M, Kawashima R, Sora I. Specific regions display altered grey matter volume in μ-opioid receptor knockout mice: MRI voxel-based morphometry. Br J Pharmacol 2014; 172:654-67. [PMID: 24913308 DOI: 10.1111/bph.12807] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 05/09/2014] [Accepted: 05/24/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE μ Opioid receptor knockout (MOP-KO) mice display several behavioural differences from wild-type (WT) littermates including differential responses to nociceptive stimuli. Brain structural changes have been tied to behavioural alterations noted in transgenic mice with targeting of different genes. Hence, we assess the brain structure of MOP-KO mice. EXPERIMENTAL APPROACH Magnetic resonance imaging (MRI) voxel-based morphometry (VBM) and histological methods were used to identify structural differences between extensively backcrossed MOP-KO mice and WT mice. KEY RESULTS MOP-KO mice displayed robust increases in regional grey matter volume in olfactory bulb, several hypothalamic nuclei, periaqueductal grey (PAG) and several cerebellar areas, most confirmed by VBM analysis. The largest increases in grey matter volume were detected in the glomerular layer of the olfactory bulb, arcuate nucleus of hypothalamus, ventrolateral PAG (VLPAG) and cerebellar regions including paramedian and cerebellar lobules. Histological analyses confirm several of these results, with increased VLPAG cell numbers and increased thickness of the olfactory bulb granule cell layer and cerebellar molecular and granular cell layers. CONCLUSIONS AND IMPLICATIONS MOP deletion causes previously undescribed structural changes in specific brain regions, but not in all regions with high MOP receptor densities (e.g. thalamus, nucleus accumbens) or that exhibit adult neurogenesis (e.g. hippocampus). Volume differences in hypothalamus and PAG may reflect behavioural changes including hyperalgesia. Although the precise relationship between volume change and MOP receptor deletion was not determined from this study alone, these findings suggest that levels of MOP receptor expression may influence a broader range of neural structure and function in humans than previously supposed. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- Kazumasu Sasaki
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Functional Brain Imaging, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|