1
|
Duan Y, Ye C, Liao J, Xie X. LY2940094, an NOPR antagonist, promotes oligodendrocyte generation and myelin recovery in an NOPR independent manner. Neurotherapeutics 2024; 21:e00424. [PMID: 39004556 DOI: 10.1016/j.neurot.2024.e00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
The myelin sheath plays crucial roles in brain development and neuronal functions. In the central nervous system, myelin is generated by oligodendrocytes, that differentiate from oligodendrocyte progenitor cells (OPC). In demyelinating diseases, the differentiation capacity of OPC is impaired and remyelination is dampened. Boosting remyelination by promoting OPC differentiation is a novel strategy for the treatment of demyelinating diseases. The opioid system, which consists of four receptors and their ligands, has been implicated in OPC differentiation and myelin formation. However, the exact roles of each opioid receptor and the relevant pharmacological molecules in OPC differentiation and myelin formation remain elusive. In the present study, specific agonists and antagonists of each opioid receptor were used to explore the function of opioid receptors in OPC differentiation. Nociceptin/orphanin FQ receptor (NOPR) specific antagonist LY2940094 was found to stimulate OPC differentiation and myelination in both in vitro and in vivo models. Unexpectedly, other NOPR ligands did not affect OPC differentiation, and NOPR knockdown did not mimic or impede the effect of LY2940094. LY2940094 was found to modulate the expression of the oligodendrocytes differentiation-associated transcription factors ID4 and Myrf, although the exact mechanism remains unclear. Since LY2940094 has been tested clinically to treat depression and alcohol dependency and has displayed an acceptable safety profile, it may provide an alternative approach to treat demyelinating diseases.
Collapse
Affiliation(s)
- Yanhui Duan
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Chenyuan Ye
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China; State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingyi Liao
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China; State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xin Xie
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China; State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China.
| |
Collapse
|
2
|
Moreno-Campos R, Singleton EW, Uribe RA. A targeted CRISPR-Cas9 mediated F0 screen identifies genes involved in establishment of the enteric nervous system. PLoS One 2024; 19:e0303914. [PMID: 38809858 PMCID: PMC11135701 DOI: 10.1371/journal.pone.0303914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/02/2024] [Indexed: 05/31/2024] Open
Abstract
The vertebrate enteric nervous system (ENS) is a crucial network of enteric neurons and glia resident within the entire gastrointestinal tract (GI). Overseeing essential GI functions such as gut motility and water balance, the ENS serves as a pivotal bidirectional link in the gut-brain axis. During early development, the ENS is primarily derived from enteric neural crest cells (ENCCs). Disruptions to ENCC development, as seen in conditions like Hirschsprung disease (HSCR), lead to the absence of ENS in the GI, particularly in the colon. In this study, using zebrafish, we devised an in vivo F0 CRISPR-based screen employing a robust, rapid pipeline integrating single-cell RNA sequencing, CRISPR reverse genetics, and high-content imaging. Our findings unveil various genes, including those encoding opioid receptors, as possible regulators of ENS establishment. In addition, we present evidence that suggests opioid receptor involvement in the neurochemical coding of the larval ENS. In summary, our work presents a novel, efficient CRISPR screen targeting ENS development, facilitating the discovery of previously unknown genes, and increasing knowledge of nervous system construction.
Collapse
Affiliation(s)
- Rodrigo Moreno-Campos
- Biosciences Department, Rice University, Houston, Texas, United States of America
- Laboratory of Neural Crest and Enteric Nervous System Development, Rice University, Houston, Texas, United States of America
| | - Eileen W. Singleton
- Biosciences Department, Rice University, Houston, Texas, United States of America
- Laboratory of Neural Crest and Enteric Nervous System Development, Rice University, Houston, Texas, United States of America
| | - Rosa A. Uribe
- Biosciences Department, Rice University, Houston, Texas, United States of America
- Laboratory of Neural Crest and Enteric Nervous System Development, Rice University, Houston, Texas, United States of America
| |
Collapse
|
3
|
Moreno-Campos R, Singleton EW, Uribe RA. A targeted CRISPR-Cas9 mediated F0 screen identifies genes involved in establishment of the enteric nervous system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.28.573581. [PMID: 38234831 PMCID: PMC10793464 DOI: 10.1101/2023.12.28.573581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The vertebrate enteric nervous system (ENS) is a crucial network of enteric neurons and glia resident within the entire gastrointestinal tract (GI). Overseeing essential GI functions such as gut motility and water balance, the ENS serves as a pivotal bidirectional link in the gut-brain axis. During early development, the ENS is primarily derived from enteric neural crest cells (ENCCs). Disruptions to ENCC development, as seen in conditions like Hirschsprung disease (HSCR), lead to absence of ENS in the GI, particularly in the colon. In this study, using zebrafish, we devised an in vivo F0 CRISPR-based screen employing a robust, rapid pipeline integrating single-cell RNA sequencing, CRISPR reverse genetics, and high-content imaging. Our findings unveil various genes, including those encoding for opioid receptors, as possible regulators of ENS establishment. In addition, we present evidence that suggests opioid receptor involvement in neurochemical coding of the larval ENS. In summary, our work presents a novel, efficient CRISPR screen targeting ENS development, facilitating the discovery of previously unknown genes, and increasing knowledge of nervous system construction.
Collapse
Affiliation(s)
- Rodrigo Moreno-Campos
- Biosciences Department, Rice University, Houston, Texas, 77005, U.S.A
- Laboratory of Neural Crest and Enteric Nervous System Development, Rice University, Houston, Texas, 77005, U.S.A
| | - Eileen W. Singleton
- Biosciences Department, Rice University, Houston, Texas, 77005, U.S.A
- Laboratory of Neural Crest and Enteric Nervous System Development, Rice University, Houston, Texas, 77005, U.S.A
| | - Rosa A. Uribe
- Biosciences Department, Rice University, Houston, Texas, 77005, U.S.A
- Laboratory of Neural Crest and Enteric Nervous System Development, Rice University, Houston, Texas, 77005, U.S.A
| |
Collapse
|
4
|
Tollefson S, Stoughton C, Himes ML, McKinney KE, Mason S, Ciccocioppo R, Narendran R. Imaging Nociceptin Opioid Peptide Receptors in Alcohol Use Disorder With [ 11C]NOP-1A and Positron Emission Tomography: Findings From a Second Cohort. Biol Psychiatry 2023; 94:416-423. [PMID: 36906499 PMCID: PMC10319913 DOI: 10.1016/j.biopsych.2022.12.022] [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: 09/06/2022] [Revised: 12/14/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Nociceptin, which binds to the nociceptin opioid peptide receptor (NOP), regulates stress and reward in addiction. In a previous [11C]NOP-1A positron emission tomography (PET) study, we found no differences in NOP in non-treatment-seeking individuals with alcohol use disorder (AUD) relative to healthy control subjects Here, we evaluated NOP in treatment-seeking individuals with AUD to document its relationship with relapse to alcohol. METHODS [11C]NOP-1A distribution volume (VT) was measured in recently abstinent individuals with AUD and healthy control subjects (n = 27/group) using an arterial input function-based kinetic analysis in brain regions that regulate reward and stress behaviors. Recent heavy drinking before PET was quantified using hair ethyl glucuronide (≥30 pg/mg was defined as heavy drinking). To document relapse, 22 subjects with AUD were followed with urine ethyl glucoronide tests (3/week) for 12 weeks after PET, where they were incentivized with money to abstain. RESULTS There were no differences in [11C]NOP-1A VT between individuals with AUD and healthy control subjects. Individuals with AUD who drank heavily before the study had significantly lower VT than those with no recent heavy drinking history. Significant negative correlations between VT and the number of drinking days and the number of drinks consumed per drinking day in the 30 days before enrollment were also present. Individuals with AUD who relapsed (and dropped out) had significantly lower VT than those who abstained for 12 weeks. CONCLUSIONS Lower NOP VT in heavy drinking AUD predicted relapse to alcohol during a 12-week follow-up period. The results of this PET study support the need to investigate medications that act at NOP to prevent relapse in individuals with AUD.
Collapse
Affiliation(s)
- Savannah Tollefson
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Clara Stoughton
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael L Himes
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kaylynn E McKinney
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Scott Mason
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
5
|
Leyden GM, Greenwood MP, Gaborieau V, Han Y, Amos CI, Brennan P, Murphy D, Davey Smith G, Richardson TG. Disentangling the aetiological pathways between body mass index and site-specific cancer risk using tissue-partitioned Mendelian randomisation. Br J Cancer 2023; 128:618-625. [PMID: 36434155 PMCID: PMC9938133 DOI: 10.1038/s41416-022-02060-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 10/30/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Body mass index (BMI) is known to influence the risk of various site-specific cancers, however, dissecting which subcomponents of this heterogenous risk factor are predominantly responsible for driving disease effects has proven difficult to establish. We have leveraged tissue-specific gene expression to separate the effects of distinct phenotypes underlying BMI on the risk of seven site-specific cancers. METHODS SNP-exposure estimates were weighted in a multivariable Mendelian randomisation analysis by their evidence for colocalization with subcutaneous adipose- and brain-tissue-derived gene expression using a recently developed methodology. RESULTS Our results provide evidence that brain-tissue-derived BMI variants are predominantly responsible for driving the genetically predicted effect of BMI on lung cancer (OR: 1.17; 95% CI: 1.01-1.36; P = 0.03). Similar findings were identified when analysing cigarettes per day as an outcome (Beta = 0.44; 95% CI: 0.26-0.61; P = 1.62 × 10-6), highlighting a possible shared aetiology or mediator effect between brain-tissue BMI, smoking and lung cancer. Our results additionally suggest that adipose-tissue-derived BMI variants may predominantly drive the effect of BMI and increased risk for endometrial cancer (OR: 1.71; 95% CI: 1.07-2.74; P = 0.02), highlighting a putatively important role in the aetiology of endometrial cancer. CONCLUSIONS The study provides valuable insight into the divergent underlying pathways between BMI and the risk of site-specific cancers.
Collapse
Affiliation(s)
- Genevieve M Leyden
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, UK.
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK.
| | - Michael P Greenwood
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - Valérie Gaborieau
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Paul Brennan
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - David Murphy
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, UK
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, UK.
| |
Collapse
|
6
|
Iusupov IR, Lukyanenko ER, Altieri A, Kurkin AV. Design and Synthesis of Fsp3-Enriched Spirocyclic-Based Biological Screening Compound Arrays via DOS Strategies and Their NNMT Inhibition Profiling. ChemMedChem 2022; 17:e202200394. [PMID: 36193863 DOI: 10.1002/cmdc.202200394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/26/2022] [Indexed: 01/14/2023]
Abstract
Medicinal chemists are keen to explore tridimensional compounds, especially when it comes to small molecules. It has already been stressed that the majority of known drugs tend to be flat, whereas natural products tend to be more tridimensional and represent a good source of active compounds. 3D metrics have been implemented and computational descriptors are available to evaluate and prioritize compounds based on their 3D geometry. This is usually done by comparing the saturated carbon atoms in a molecule with the total number of its non-hydrogen atoms (the Fsp3 value). While this aspect is clear, still there are not enough synthetic tools that support the realization of novel chemotypes that conform to these criteria. Herein we describe a diversity oriented synthesis (DOS) synthetic cascade technology that starts from two simple reagents, and generates highly enriched Fsp3 novel and diverse spiro-scaffolds with pragmatic synthetic handles (points of diversity). The spiro nature of these scaffolds not only ensures high Fsp3 values but renders the compounds more rigid and therefore more effective in forming precise stereo-interactions with their potential biological targets. These compounds were also profiled for their drug-like properties and as potential modulators of the NNMT enzyme.
Collapse
Affiliation(s)
- Ildar R Iusupov
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, Moscow, 119991, Russia
| | - Evgeny R Lukyanenko
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, Moscow, 119991, Russia
| | - Andrea Altieri
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, Moscow, 119991, Russia.,EDASA Scientific Srls, Via Stingi, 3, 66050, San Salvo, Italy
| | - Alexander V Kurkin
- Department of Chemistry, Lomonosov Moscow State University, 1/3 Leninsky Gory, Moscow, 119991, Russia
| |
Collapse
|
7
|
Brain serotonin deficiency and fluoxetine lead to sex-specific effects on binge-like food consumption in mice. Psychopharmacology (Berl) 2022; 239:2975-2984. [PMID: 35750862 DOI: 10.1007/s00213-022-06181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
RATIONALE Although pharmacotherapies are often effective in reducing binge eating in conditions such as bulimia nervosa and binge eating disorder, subsets of patients do not benefit sufficiently from existing treatments, and the reasons for treatment failure remain unclear. OBJECTIVES This study aimed to evaluate whether genetic reductions in brain serotonin influence binge eating and/or the ability of fluoxetine, a selective serotonin reuptake inhibitor, to reduce binge eating in mice. METHODS This study used a validated model of binge-like consumption of high-fat diet to compare binge-like food intake in control and fluoxetine-treated wild-type and serotonin-deficient mice from the tryptophan hydroxylase 2 (R439H) knock-in line. In addition, real-time PCR was used to evaluate potential genotype and sex differences in the effects of fluoxetine on gene expression in the raphe nucleus. RESULTS The results reveal that brain serotonin deficiency is sufficient to increase binge eating in males, but not females. However, while chronic fluoxetine reduced binge eating in both genotypes of males and in wild-type females, it failed to reduce binge eating in serotonin-deficient females. Transcriptional responses to chronic fluoxetine were also characterized by sex and genotype differences. CONCLUSIONS Overall, this study revealed significant sex differences in the effects of fluoxetine and brain serotonin deficiency on binge-like food intake and suggests that low brain serotonin could impact eating disorders both by promoting binge eating and by limiting the efficacy of fluoxetine to reduce binge eating.
Collapse
|
8
|
Heal DJ, Smith SL. Prospects for new drugs to treat binge-eating disorder: Insights from psychopathology and neuropharmacology. J Psychopharmacol 2022; 36:680-703. [PMID: 34318734 PMCID: PMC9150143 DOI: 10.1177/02698811211032475] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Binge-eating disorder (BED) is a common psychiatric condition with adverse psychological and metabolic consequences. Lisdexamfetamine (LDX) is the only approved BED drug treatment. New drugs to treat BED are urgently needed. METHODS A comprehensive review of published psychopathological, pharmacological and clinical findings. RESULTS The evidence supports the hypothesis that BED is an impulse control disorder with similarities to ADHD, including responsiveness to catecholaminergic drugs, for example LDX and dasotraline. The target product profile (TPP) of the ideal BED drug combines treating the psychopathological drivers of the disorder with an independent weight-loss effect. Drugs with proven efficacy in BED have a common pharmacology; they potentiate central noradrenergic and dopaminergic neurotransmission. Because of the overlap between pharmacotherapy in attention deficit hyperactivity disorder (ADHD) and BED, drug-candidates from diverse pharmacological classes, which have already failed in ADHD would also be predicted to fail if tested in BED. The failure in BED trials of drugs with diverse pharmacological mechanisms indicates many possible avenues for drug discovery can probably be discounted. CONCLUSIONS (1) The efficacy of drugs for BED is dependent on reducing its core psychopathologies of impulsivity, compulsivity and perseveration and by increasing cognitive control of eating. (2) The analysis revealed a large number of pharmacological mechanisms are unlikely to be productive in the search for effective new BED drugs. (3) The most promising areas for new treatments for BED are drugs, which augment noradrenergic and dopaminergic neurotransmission and/or those which are effective in ADHD.
Collapse
Affiliation(s)
- David J Heal
- David J Heal, DevelRx Ltd, BioCity, Nottingham, NG1 1GF, UK.
| | | |
Collapse
|
9
|
D'Oliveira da Silva F, Azevedo Neto J, Sturaro C, Guarino A, Robert C, Gavioli EC, Calo G, Mouledous L, Ruzza C. The NOP antagonist BTRX-246040 increases stress resilience in mice without affecting adult neurogenesis in the hippocampus. Neuropharmacology 2022; 212:109077. [DOI: 10.1016/j.neuropharm.2022.109077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/04/2022] [Accepted: 04/27/2022] [Indexed: 11/24/2022]
|
10
|
Ubaldi M, Cannella N, Borruto AM, Petrella M, Micioni Di Bonaventura MV, Soverchia L, Stopponi S, Weiss F, Cifani C, Ciccocioppo R. Role of Nociceptin/Orphanin FQ-NOP Receptor System in the Regulation of Stress-Related Disorders. Int J Mol Sci 2021; 22:12956. [PMID: 34884757 PMCID: PMC8657682 DOI: 10.3390/ijms222312956] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022] Open
Abstract
Nociceptin/orphanin FQ (N/OFQ) is a 17-residue neuropeptide that binds the nociceptin opioid-like receptor (NOP). N/OFQ exhibits nucleotidic and aminoacidics sequence homology with the precursors of other opioid neuropeptides but it does not activate either MOP, KOP or DOP receptors. Furthermore, opioid neuropeptides do not activate the NOP receptor. Generally, activation of N/OFQ system exerts anti-opioids effects, for instance toward opioid-induced reward and analgesia. The NOP receptor is widely expressed throughout the brain, whereas N/OFQ localization is confined to brain nuclei that are involved in stress response such as amygdala, BNST and hypothalamus. Decades of studies have delineated the biological role of this system demonstrating its involvement in significant physiological processes such as pain, learning and memory, anxiety, depression, feeding, drug and alcohol dependence. This review discusses the role of this peptidergic system in the modulation of stress and stress-associated psychiatric disorders in particular drug addiction, mood, anxiety and food-related associated-disorders. Emerging preclinical evidence suggests that both NOP agonists and antagonists may represent a effective therapeutic approaches for substances use disorder. Moreover, the current literature suggests that NOP antagonists can be useful to treat depression and feeding-related diseases, such as obesity and binge eating behavior, whereas the activation of NOP receptor by agonists could be a promising tool for anxiety.
Collapse
Affiliation(s)
- Massimo Ubaldi
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Nazzareno Cannella
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Anna Maria Borruto
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Michele Petrella
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Maria Vittoria Micioni Di Bonaventura
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Laura Soverchia
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Serena Stopponi
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA;
| | - Carlo Cifani
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| | - Roberto Ciccocioppo
- School of Pharmacy, University of Camerino, Via Madonna Delle Carceri 9, 62032 Camerino, Italy; (M.U.); (N.C.); (A.M.B.); (M.P.); (M.V.M.D.B.); (L.S.); (S.S.); (C.C.)
| |
Collapse
|
11
|
Gastelum C, Perez L, Hernandez J, Le N, Vahrson I, Sayers S, Wagner EJ. Adaptive Changes in the Central Control of Energy Homeostasis Occur in Response to Variations in Energy Status. Int J Mol Sci 2021; 22:2728. [PMID: 33800452 PMCID: PMC7962960 DOI: 10.3390/ijms22052728] [Citation(s) in RCA: 12] [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: 02/05/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/15/2022] Open
Abstract
Energy homeostasis is regulated in coordinate fashion by the brain-gut axis, the homeostatic energy balance circuitry in the hypothalamus and the hedonic energy balance circuitry comprising the mesolimbcortical A10 dopamine pathway. Collectively, these systems convey and integrate information regarding nutrient status and the rewarding properties of ingested food, and formulate it into a behavioral response that attempts to balance fluctuations in consumption and food-seeking behavior. In this review we start with a functional overview of the homeostatic and hedonic energy balance circuitries; identifying the salient neural, hormonal and humoral components involved. We then delve into how the function of these circuits differs in males and females. Finally, we turn our attention to the ever-emerging roles of nociceptin/orphanin FQ (N/OFQ) and pituitary adenylate cyclase-activating polypeptide (PACAP)-two neuropeptides that have garnered increased recognition for their regulatory impact in energy homeostasis-to further probe how the imposed regulation of energy balance circuitry by these peptides is affected by sex and altered under positive (e.g., obesity) and negative (e.g., fasting) energy balance states. It is hoped that this work will impart a newfound appreciation for the intricate regulatory processes that govern energy homeostasis, as well as how recent insights into the N/OFQ and PACAP systems can be leveraged in the treatment of conditions ranging from obesity to anorexia.
Collapse
Affiliation(s)
- Cassandra Gastelum
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Lynnea Perez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Jennifer Hernandez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Nikki Le
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Isabella Vahrson
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Sarah Sayers
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Edward J. Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
12
|
Micioni Di Bonaventura E, Botticelli L, Tomassoni D, Tayebati SK, Micioni Di Bonaventura MV, Cifani C. The Melanocortin System behind the Dysfunctional Eating Behaviors. Nutrients 2020; 12:E3502. [PMID: 33202557 PMCID: PMC7696960 DOI: 10.3390/nu12113502] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
The dysfunction of melanocortin signaling has been associated with obesity, given the important role in the regulation of energy homeostasis, food intake, satiety and body weight. In the hypothalamus, the melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R) contribute to the stability of these processes, but MC3R and MC4R are also localized in the mesolimbic dopamine system, the region that responds to the reinforcing properties of highly palatable food (HPF) and where these two receptors seem to affect food reward and motivation. Loss of function of the MC4R, resulting from genetic mutations, leads to overeating in humans, but to date, a clear understanding of the underlying mechanisms and behaviors that promote overconsumption of caloric foods remains unknown. Moreover, the MC4R demonstrated to be a crucial modulator of the stress response, factor that is known to be strictly related to binge eating behavior. In this review, we will explore the preclinical and clinical studies, and the controversies regarding the involvement of melanocortin system in altered eating patterns, especially binge eating behavior, food reward and motivation.
Collapse
Affiliation(s)
| | - Luca Botticelli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | - Daniele Tomassoni
- School of Bioscience and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy;
| | - Seyed Khosrow Tayebati
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | | | - Carlo Cifani
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| |
Collapse
|
13
|
Jais A, Paeger L, Sotelo-Hitschfeld T, Bremser S, Prinzensteiner M, Klemm P, Mykytiuk V, Widdershooven PJM, Vesting AJ, Grzelka K, Minère M, Cremer AL, Xu J, Korotkova T, Lowell BB, Zeilhofer HU, Backes H, Fenselau H, Wunderlich FT, Kloppenburg P, Brüning JC. PNOC ARC Neurons Promote Hyperphagia and Obesity upon High-Fat-Diet Feeding. Neuron 2020; 106:1009-1025.e10. [PMID: 32302532 PMCID: PMC7303947 DOI: 10.1016/j.neuron.2020.03.022] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/11/2020] [Accepted: 03/23/2020] [Indexed: 12/30/2022]
Abstract
Calorie-rich diets induce hyperphagia and promote obesity, although the underlying mechanisms remain poorly defined. We find that short-term high-fat-diet (HFD) feeding of mice activates prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus of the hypothalamus (ARC). PNOCARC neurons represent a previously unrecognized GABAergic population of ARC neurons distinct from well-defined feeding regulatory AgRP or POMC neurons. PNOCARC neurons arborize densely in the ARC and provide inhibitory synaptic input to nearby anorexigenic POMC neurons. Optogenetic activation of PNOCARC neurons in the ARC and their projections to the bed nucleus of the stria terminalis promotes feeding. Selective ablation of these cells promotes the activation of POMC neurons upon HFD exposure, reduces feeding, and protects from obesity, but it does not affect food intake or body weight under normal chow consumption. We characterize PNOCARC neurons as a novel ARC neuron population activated upon palatable food consumption to promote hyperphagia. Acute high-fat-diet feeding activates PNOC neurons in the arcuate nucleus (ARC) GABAergic PNOCARC neurons inhibit anorexigenic POMC neurons Optogenetic activation of PNOCARC neurons promotes feeding Ablation of PNOCARC neurons protects from obesity
Collapse
Affiliation(s)
- Alexander Jais
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Lars Paeger
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Tamara Sotelo-Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Stephan Bremser
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany
| | - Melanie Prinzensteiner
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Vasyl Mykytiuk
- Neuronal Circuits and Behaviour Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Pia J M Widdershooven
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Anna Juliane Vesting
- Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Obesity and Cancer Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Katarzyna Grzelka
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Marielle Minère
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Anna Lena Cremer
- Multimodal Imaging of Brain Metabolism Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Jie Xu
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Tatiana Korotkova
- Neuronal Circuits and Behaviour Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - Heiko Backes
- Multimodal Imaging of Brain Metabolism Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Henning Fenselau
- Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - F Thomas Wunderlich
- Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Obesity and Cancer Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Institute for Zoology, Biocenter, University of Cologne, Cologne, Germany.
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
14
|
Borruto AM, Fotio Y, Stopponi S, Brunori G, Petrella M, Caputi FF, Romualdi P, Candeletti S, Narendran R, Rorick-Kehn LM, Ubaldi M, Weiss F, Ciccocioppo R. NOP receptor antagonism reduces alcohol drinking in male and female rats through mechanisms involving the central amygdala and ventral tegmental area. Br J Pharmacol 2020; 177:1525-1537. [PMID: 31713848 DOI: 10.1111/bph.14915] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Nociceptin/orphanin FQ (N/OFQ) peptide and its cognate receptor (NOP) are widely expressed in mesolimbic brain regions where they play an important role in modulating reward and motivation. Early evidence suggested that NOP receptor activation attenuates the rewarding effects of drugs of abuse, including alcohol. However, emerging data indicate that NOP receptor blockade also effectively attenuates alcohol drinking and relapse. To advance our understanding of the role of the N/OFQ-NOP receptor system in alcohol abuse, we examined the effect of NOP receptor blockade on voluntary alcohol drinking at the neurocircuitry level. EXPERIMENTAL APPROACH Using male and female genetically selected alcohol-preferring Marchigian Sardinian (msP) rats, we initially evaluated the effects of the selective NOP receptor antagonist LY2817412 (3, 10, and 30 mg·kg-1 , p.o.) on alcohol consumption in a two-bottle free-choice paradigm. We then microinjected LY2817412 (3 and 6 μg·μl-1 per rat) in the central nucleus of the amygdala (CeA), ventral tegmental area (VTA), and nucleus accumbens (NAc). KEY RESULTS Peripheral LY2817412 administration dose-dependently and selectively reduced voluntary alcohol intake in male and female msP rats. Central injections of LY2817412 markedly attenuated voluntary alcohol intake in both sexes following administration in the CeA and VTA but not in the NAc. CONCLUSION AND IMPLICATIONS The present results revealed that the CeA and VTA are neuroanatomical substrates that mediate the effects of NOP receptor antagonism on alcohol consumption. Overall, our findings support the potential of NOP receptor antagonism as a treatment strategy to attenuate alcohol use and addiction.
Collapse
Affiliation(s)
| | - Yannick Fotio
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Gloria Brunori
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy.,Department of Biomedical Science, Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Michele Petrella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Francesca Felicia Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Linda M Rorick-Kehn
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Italy
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
15
|
Jewett DC, Klockars A, Smith TR, Brunton C, Head MA, Tham RL, Kwilasz AJ, Hahn TW, Wiebelhaus JM, Ewan EE, Carroll RM, Grace MK, Levine AS, Olszewski PK. Effects of opioid receptor ligands in rats trained to discriminate 22 from 2 hours of food deprivation suggest a lack of opioid involvement in eating for hunger. Behav Brain Res 2020; 380:112369. [DOI: 10.1016/j.bbr.2019.112369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/28/2019] [Accepted: 11/15/2019] [Indexed: 10/25/2022]
|
16
|
Ferrari F, Rizzo S, Ruzza C, Calo G. Detailed In Vitro Pharmacological Characterization of the Clinically Viable Nociceptin/Orphanin FQ Peptide Receptor Antagonist BTRX-246040. J Pharmacol Exp Ther 2020; 373:34-43. [PMID: 31937563 DOI: 10.1124/jpet.119.262865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/07/2020] [Indexed: 12/20/2022] Open
Abstract
The peptide nociceptin/orphanin FQ (N/OFQ) is the natural ligand of the N/OFQ receptor (NOP), which is widely expressed in the central and peripheral nervous system. Selective NOP antagonists are worthy of testing as innovative drugs to treat depression, Parkinson disease, and drug abuse. The aim of this study was to perform a detailed in vitro characterization of BTRX-246040 (also known as LY2940094, [2-[4-[(2-chloro-4,4-difluoro-spiro[5H-thieno[2,3-c]pyran-7,4'-piperidine]-1'-yl)methyl]-3-methyl-pyrazol-1-yl]-3-pyridyl]methanol), a novel NOP antagonist that has been already studied in humans. BTRX-246040 has been tested in vitro in the following assays: calcium mobilization in cells expressing NOP and classic opioid receptors and chimeric G proteins, bioluminescence resonance energy transfer assay measuring NOP interaction with G proteins and β-arrestins, the label-free dynamic mass redistribution assay, and the electrically stimulated mouse vas deferens. BTRX-246040 was systematically compared with the standard NOP antagonist SB-612111. In all assays, BTRX-246040 behaves as a pure and selective antagonist at human recombinant and murine native NOP receptors displaying 3-10-fold higher potency than the standard antagonist SB-612111. BTRX-246040 is an essential pharmacological tool to further investigate the therapeutic potential of NOP antagonists in preclinical and clinical studies. SIGNIFICANCE STATEMENT: NOP antagonists may be innovative antidepressant drugs. In this research, the novel clinically viable NOP antagonist BTRX-246040 has been deeply characterized in vitro in a panel of assays. BTRX-246040 resulted a pure, potent, and selective NOP antagonist.
Collapse
Affiliation(s)
- Federica Ferrari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy (F.F., S.R., C.R., G.C.) and Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, Ferrara, Italy (C.R.)
| | - Sabrina Rizzo
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy (F.F., S.R., C.R., G.C.) and Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, Ferrara, Italy (C.R.)
| | - Chiara Ruzza
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy (F.F., S.R., C.R., G.C.) and Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, Ferrara, Italy (C.R.)
| | - Girolamo Calo
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy (F.F., S.R., C.R., G.C.) and Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, Ferrara, Italy (C.R.)
| |
Collapse
|
17
|
Brunori G, Weger M, Schoch J, Targowska-Duda K, Barnes M, Borruto AM, Rorick-Kehn LM, Zaveri NT, Pintar JE, Ciccocioppo R, Toll L, Cippitelli A. NOP Receptor Antagonists Decrease Alcohol Drinking in the Dark in C57BL/6J Mice. Alcohol Clin Exp Res 2019; 43:2167-2178. [PMID: 31386211 DOI: 10.1111/acer.14165] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The nociceptin/orphanin FQ opioid peptide (NOP) receptor and its endogenous ligand N/OFQ have been implicated in the regulation of drug and alcohol use disorders (AUD). In particular, evidence demonstrated that NOP receptor activation blocks reinforcing and motivating effects of alcohol across a range of behavioral measures, including alcohol intake, conditioned place preference, and vulnerability to relapse. METHODS Here, we show the effects of pharmacological activation and inhibition of NOP receptors on binge-like alcohol consumption, as measured by the "drinking in the dark" (DID) model in C57BL/6J mice. RESULTS We found that 2 potent and selective NOP agonists AT-202 (0, 0.3, 1, 3 mg/kg) and AT-312 (0, 0.3, 1 mg/kg) did not affect binge alcohol drinking at doses that do not affect locomotor activity. AT-202 also failed to alter DID behavior when administered to mice previously exposed to chronic alcohol treatment with an alcohol-containing liquid diet. Conversely, treatment with either the high affinity NOP receptor antagonist SB-612111 (0, 3, 10, 30 mg/kg) or the selective antagonist LY2817412 (0, 3, 10, 30 mg/kg) decreased binge drinking. SB-612111 was effective at all doses examined, and LY2817412 was effective at 30 mg/kg. Consistently, NOP receptor knockout mice consumed less alcohol compared to wild type. SB-612111 reduced DID and increased sucrose consumption at doses that do not appear to affect locomotor activity. However, the high dose of SB-612111 (30 mg/kg) reduced alcohol intake but failed to inhibit preference in a 2-bottle choice DID model that can assess moderate alcohol intake. CONCLUSIONS The present results suggest that NOP receptor inhibition rather than activation may represent a valuable approach for treatment of AUD characterized by excessive alcohol consumption such as binge drinking.
Collapse
Affiliation(s)
- Gloria Brunori
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida.,Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida.,Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Michelle Weger
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Jennifer Schoch
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida.,Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Katarzyna Targowska-Duda
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida.,Department of Biopharmacy, Medical University of Lublin, Lublin, Poland
| | - Megan Barnes
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida
| | - Anna Maria Borruto
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | | | | | - John E Pintar
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Lawrence Toll
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida.,Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| | - Andrea Cippitelli
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida.,Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida
| |
Collapse
|
18
|
Bodnar RJ. Endogenous opioid modulation of food intake and body weight: Implications for opioid influences upon motivation and addiction. Peptides 2019; 116:42-62. [PMID: 31047940 DOI: 10.1016/j.peptides.2019.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 03/04/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
This review is part of a special issue dedicated to Opioid addiction, and examines the influential role of opioid peptides, opioid receptors and opiate drugs in mediating food intake and body weight control in rodents. This review postulates that opioid mediation of food intake was an example of "positive addictive" properties that provide motivational drives to maintain opioid-seeking behavior and that are not subject to the "negative addictive" properties associated with tolerance, dependence and withdrawal. Data demonstrate that opiate and opioid peptide agonists stimulate food intake through homeostatic activation of sensory, metabolic and energy-related In contrast, general, and particularly mu-selective, opioid receptor antagonists typically block these homeostatically-driven ingestive behaviors. Intake of palatable and hedonic food stimuli is inhibited by general, and particularly mu-selective, opioid receptor antagonists. The selectivity of specific opioid agonists to elicit food intake was confirmed through the use of opioid receptor antagonists and molecular knockdown (antisense) techniques incapacitating specific exons of opioid receptor genes. Further extensive evidence demonstrated that homeostatic and hedonic ingestive situations correspondingly altered the levels and expression of opioid peptides and opioid receptors. Opioid mediation of food intake was controlled by a distributed brain network intimately related to both the appetitive-consummatory sites implicated in food intake as well as sites intimately involved in reward and reinforcement. This emergent system appears to sustain the "positive addictive" properties providing motivational drives to maintain opioid-seeking behavior.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology, Queens College, City University of New York, United States; Psychology Doctoral Program and CUNY Neuroscience Collaborative, The Graduate Center of the City University of New York, United States.
| |
Collapse
|
19
|
Browne CA, Lucki I. Targeting opioid dysregulation in depression for the development of novel therapeutics. Pharmacol Ther 2019; 201:51-76. [PMID: 31051197 DOI: 10.1016/j.pharmthera.2019.04.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
Since the serendipitous discovery of the first class of modern antidepressants in the 1950's, all pharmacotherapies approved by the Food and Drug Administration for major depressive disorder (MDD) have shared a common mechanism of action, increased monoaminergic neurotransmission. Despite the widespread availability of antidepressants, as many as 50% of depressed patients are resistant to these conventional therapies. The significant length of time required to produce meaningful symptom relief with these medications, 4-6 weeks, indicates that other mechanisms are likely involved in the pathophysiology of depression which may yield more viable targets for drug development. For decades, no viable candidate target with a different mechanism of action to that of conventional therapies proved successful in clinical studies. Now several exciting avenues for drug development are under intense investigation. One of these emerging targets is modulation of endogenous opioid tone. This review will evaluate preclinical and clinical evidence pertaining to opioid dysregulation in depression, focusing on the role of the endogenous ligands endorphin, enkephalin, dynorphin, and nociceptin/orphanin FQ (N/OFQ) and their respective receptors, mu (MOR), delta (DOR), kappa (KOR), and the N/OFQ receptor (NOP) in mediating behaviors relevant to depression and anxiety. Finally, putative opioid based antidepressants that are under investigation in clinical trials, ALKS5461, JNJ-67953964 (formerly LY2456302 and CERC-501) and BTRX-246040 (formerly LY-2940094) will be discussed. This review will illustrate the potential therapeutic value of targeting opioid dysregulation in developing novel therapies for MDD.
Collapse
Affiliation(s)
- Caroline A Browne
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America
| | - Irwin Lucki
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| |
Collapse
|
20
|
Hardaway JA, Halladay LR, Mazzone CM, Pati D, Bloodgood DW, Kim M, Jensen J, DiBerto JF, Boyt KM, Shiddapur A, Erfani A, Hon OJ, Neira S, Stanhope CM, Sugam JA, Saddoris MP, Tipton G, McElligott Z, Jhou TC, Stuber GD, Bruchas MR, Bulik CM, Holmes A, Kash TL. Central Amygdala Prepronociceptin-Expressing Neurons Mediate Palatable Food Consumption and Reward. Neuron 2019; 102:1037-1052.e7. [PMID: 31029403 DOI: 10.1016/j.neuron.2019.03.037] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 09/27/2018] [Accepted: 03/27/2019] [Indexed: 01/04/2023]
Abstract
Food palatability is one of many factors that drives food consumption, and the hedonic drive to feed is a key contributor to obesity and binge eating. In this study, we identified a population of prepronociceptin-expressing cells in the central amygdala (PnocCeA) that are activated by palatable food consumption. Ablation or chemogenetic inhibition of these cells reduces palatable food consumption. Additionally, ablation of PnocCeA cells reduces high-fat-diet-driven increases in bodyweight and adiposity. PnocCeA neurons project to the ventral bed nucleus of the stria terminalis (vBNST), parabrachial nucleus (PBN), and nucleus of the solitary tract (NTS), and activation of cell bodies in the central amygdala (CeA) or axons in the vBNST, PBN, and NTS produces reward behavior but did not promote feeding of palatable food. These data suggest that the PnocCeA network is necessary for promoting the reinforcing and rewarding properties of palatable food, but activation of this network itself is not sufficient to promote feeding.
Collapse
Affiliation(s)
- J Andrew Hardaway
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA.
| | - Lindsay R Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA; Department of Psychology, Santa Clara University, Santa Clara, CA 95053, USA
| | - Christopher M Mazzone
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dipanwita Pati
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Daniel W Bloodgood
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michelle Kim
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Jennifer Jensen
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Jeffrey F DiBerto
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Kristen M Boyt
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Ami Shiddapur
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Ava Erfani
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Olivia J Hon
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sofia Neira
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christina M Stanhope
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Jonathan A Sugam
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Michael P Saddoris
- Department of Psychology and Neuroscience, University of Colorado, Boulder, Boulder, CO 80309, USA
| | - Greg Tipton
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Zoe McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Thomas C Jhou
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Garret D Stuber
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Michael R Bruchas
- Division of Basic Research, Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
21
|
Turan N, Özkay ÜD, Can NÖ, Can ÖD. Investigating the Antidepressant-like Effects of some Benzimidazolepiperidine Derivatives by In-Vivo Experimental Methods. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666181004103112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background: Benzimidazole and piperidine rings are important pharmacophore groups
for drug design studies.
</P><P>
Objective: In this study, we aimed to investigate the antidepressant-like activity of some 2-(4-
substituted-phenyl)-1-[2-(piperidin-1-yl)ethyl]-1H-benzimidazole derivatives.
</P><P>
Methods: Tail-suspension Test (TST) and Modified Forced Swimming Tests (MFST) were used to
assess antidepressant-like activities of the test compounds. Moreover, locomotor activity performances
of the animals were evaluated by an activity cage device.
</P><P>
Results: In the TST and MFST, compounds 2c-2h (10 mg/kg) and the reference drug fluoxetine (20
mg/kg) significantly reduced the immobility time of mice indicating the antidepressant-like activities
of these compounds. Further, in MFST, the same compounds induced significant enhancement
in the duration of active swimming behaviors without affecting the climbing performance of the
animals. This prolongation in the swimming time, similar to fluoxetine, pointed out that antidepressant-
like activity of the compounds 2c-2h might be related to the serotonergic rather than noradrenergic
mechanisms. Besides, results of the activity cage tests demonstrated that none of the tested
compounds caused an alteration in the locomotor activities of mice, signifying that antidepressantlike
effects presented in this study were specific.
</P><P>
Conclusion: In conclusion, results of this present study supported the previous papers reporting the
therapeutic potential of compounds carrying benzimidazole and/or piperidine rings in their structure
and emphasized, once again, the importance of these pharmacophore groups in drug design studies.
Collapse
Affiliation(s)
- Nazlı Turan
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| | - Ümide Demir Özkay
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| | - Nafiz Öncü Can
- Department of Analytical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| | - Özgür Devrim Can
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| |
Collapse
|
22
|
Witkin JM, Wallace TL, Martin WJ. Therapeutic Approaches for NOP Receptor Antagonists in Neurobehavioral Disorders: Clinical Studies in Major Depressive Disorder and Alcohol Use Disorder with BTRX-246040 (LY2940094). Handb Exp Pharmacol 2019; 254:399-415. [PMID: 30701317 DOI: 10.1007/164_2018_186] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Conventional antidepressants increase the efflux of biogenic amine neurotransmitters (the monoamine hypothesis of depression) in the central nervous system (CNS) and are the principle drugs used to treat major depressive disorder (MDD). However, the lack of efficacy in some patients, the slow onset of action, and the side effect profiles of existing antidepressants necessitate the exploration of additional treatment options. The discovery of the nociceptin/orphanin FQ peptide NOP receptor (N/OFQ-NOP receptor) system and its characterization in preclinical biological and pharmacological stress-related conditions supports the potential antidepressant and anti-stress properties of a NOP receptor antagonist for the treatment of neurobehavioral disorders. BTRX-246040 (formerly LY2940094) was designed to test this hypothesis in the clinic. A small clinical proof of concept study demonstrated efficacy of BTRX-246040 in MDD patients. In this study, BTRX-246040 (40 mg, p.o.) significantly reduced negative bias as assessed by the facial recognition test within 1 week of treatment and decreased depression symptoms after 8 weeks. BTRX-246040 also reduced depression symptoms in a second trial with heavy alcohol drinkers. Given the comorbidity of MDD and alcohol use disorder, a compound with such effects in patients could be a valuable addition to the medications available. A proof of concept study showed efficacy of BTRX-246040 in reducing heavy drinking and increasing the probability of abstinence in individuals diagnosed with alcohol dependence. In addition, plasma levels of gamma-glutamyl transferase were decreased by BTRX-246040 compared to placebo control implying improvement in liver function. Collectively, the clinical data reviewed within this chapter suggest that BTRX-264040 functions to normalize dysfunction in reward circuits. The overall efficacy and safety of this compound with a novel mechanism of action are encouraging of further clinical development. BTRX-246040 is currently under development for MDD by BlackThorn Therapeutics.
Collapse
|
23
|
McElroy SL, Guerdjikova AI, Mori N, Romo-Nava F. Progress in Developing Pharmacologic Agents to Treat Bulimia Nervosa. CNS Drugs 2019; 33:31-46. [PMID: 30523523 DOI: 10.1007/s40263-018-0594-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This paper reviews past and current progress in developing pharmacologic agents for the treatment of individuals with bulimia nervosa (BN). We searched the literature and clinical trial registries for compounds studied in BN, the related condition, binge eating disorder (BED), and preclinical models of binge-eating behavior. Drug classes evaluated included antidepressants, antiepileptic drugs, stimulants and other medications for attention-deficit/hyperactivity disorder, opioid antagonists, and weight loss agents, among others. The only available drugs with established efficacy in BN at this time include antidepressants (especially selective serotonin reuptake inhibitors [SSRIs]) and the antiepileptic topiramate, though the efficacy of these compounds is modest at best. The only medications we found currently receiving empirical study in people with BN were fluoxetine, other serotonergic antidepressants, intranasal naloxone, lisdexamfetamine dimesylate, phentermine-topiramate combination, the antiandrogenic oral contraceptive ethinyl estradiol plus drospirenone, and prazosin. Preclinical models suggest that nociceptin receptor antagonists, the selective serotonin 5-HT2C receptor agonist lorcaserin, monoamine stabilizers, and selective orexin-1 receptor antagonists might be helpful. We found no evidence of a drug developed specifically for the treatment of individuals with BN. Future areas for research in the pharmacotherapy of BN are suggested. Importantly, until drugs are developed specifically for eating disorders, drugs developed for other conditions that are centrally acting and associated with beneficial psychotropic effects and/or reduced appetite or weight loss might be considered for repurposing in BN.
Collapse
Affiliation(s)
- Susan L McElroy
- Lindner Center of HOPE, Mason, OH, USA. .,University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Anna I Guerdjikova
- Lindner Center of HOPE, Mason, OH, USA.,University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Francisco Romo-Nava
- Lindner Center of HOPE, Mason, OH, USA.,University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
24
|
Micioni Di Bonaventura MV, Micioni Di Bonaventura E, Cifani C, Polidori C. N/OFQ-NOP System in Food Intake. Handb Exp Pharmacol 2019; 254:279-295. [PMID: 31073870 DOI: 10.1007/164_2019_212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
While lifestyle modifications should be the first-line actions in preventing and treating obesity and eating disorders, pharmacotherapy also provides a necessary tool for the management of these diseases.However, given the limitations of current anti-obesity drugs, innovative treatments that improve efficacy and safety are needed.Since the discovery that the activation of the Nociceptin/Orphanin (N/OFQ) FQ peptide (NOP) receptor by N/OFQ induces an increase of food intake in laboratory animals, and the finding that this effect can be blocked by NOP antagonists, many NOP agonists and antagonists have been synthesized and tested in vitro and in vivo for their potential regulation of feeding behavior. Promising results seem to suggest that the N/OFQergic system may be a potential therapeutic target for the neural control of feeding behavior and related pathologies, especially in binge-like eating behavior.
Collapse
Affiliation(s)
| | | | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, MC, Italy.
| | - Carlo Polidori
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, MC, Italy
| |
Collapse
|
25
|
Caputi FF, Romualdi P, Candeletti S. Regulation of the Genes Encoding the ppN/OFQ and NOP Receptor. Handb Exp Pharmacol 2019; 254:141-162. [PMID: 30689088 DOI: 10.1007/164_2018_196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Over the years, the ability of N/OFQ-NOP receptor system in modulating several physiological functions, including the release of neurotransmitters, anxiety-like behavior responses, modulation of the reward circuitry, inflammatory signaling, nociception, and motor function, has been examined in several brain regions and at spinal level. This chapter collects information related to the genes encoding the ppN/OFQ and NOP receptor, their regulation, and relative transcriptional control mechanisms. Furthermore, genetic manipulations, polymorphisms, and epigenetic alterations associated with different pathological conditions are discussed. The evidence here collected indicates that the study of ppN/OFQ and NOP receptor gene expression may offer novel opportunities in the field of personalized therapies and highlights this system as a good "druggable target" for different pathological conditions.
Collapse
Affiliation(s)
- Francesca Felicia Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Corbière A, Walet-Balieu ML, Chan P, Basille-Dugay M, Hardouin J, Vaudry D. A Peptidomic Approach to Characterize Peptides Involved in Cerebellar Cortex Development Leads to the Identification of the Neurotrophic Effects of Nociceptin. Mol Cell Proteomics 2018; 17:1737-1749. [PMID: 29895708 PMCID: PMC6126386 DOI: 10.1074/mcp.ra117.000184] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 05/16/2018] [Indexed: 12/20/2022] Open
Abstract
The cerebellum is a brain structure involved in motor and cognitive functions. The development of the cerebellar cortex (the external part of the cerebellum) is under the control of numerous factors. Among these factors, neuropeptides including PACAP or somatostatin modulate the survival, migration and/or differentiation of cerebellar granule cells. Interestingly, such peptides contributing to cerebellar ontogenesis usually exhibit a specific transient expression profile with a low abundance at birth, a high expression level during the developmental processes, which take place within the first two postnatal weeks in rodents, and a gradual decline toward adulthood. Thus, to identify new peptides transiently expressed in the cerebellum during development, rat cerebella were sampled from birth to adulthood, and analyzed by a semi-quantitative peptidomic approach. A total of 33 peptides were found to be expressed in the cerebellum. Among these 33 peptides, 8 had a clear differential expression pattern during development, 4 of them i.e. cerebellin 2, nociceptin, somatostatin and VGF [353-372], exhibiting a high expression level during the first two postnatal weeks followed by a significative decrease at adulthood. A focus by a genomic approach on nociceptin, confirmed that its precursor mRNA is transiently expressed during the first week of life in granule neurons within the internal granule cell layer of the cerebellum, and showed that the nociceptin receptor is also actively expressed between P8 and P16 by the same neurons. Finally, functional studies revealed a new role for nociceptin, acting as a neurotrophic peptide able to promote the survival and differentiation of developing cerebellar granule neurons.
Collapse
Affiliation(s)
- Auriane Corbière
- From the ‡Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, 76000 Rouen, France
| | - Marie-Laure Walet-Balieu
- §Normandie Univ, UNIROUEN, Rouen Proteomic Platform (PISSARO), Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Philippe Chan
- §Normandie Univ, UNIROUEN, Rouen Proteomic Platform (PISSARO), Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Magali Basille-Dugay
- From the ‡Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, 76000 Rouen, France
| | - Julie Hardouin
- §Normandie Univ, UNIROUEN, Rouen Proteomic Platform (PISSARO), Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - David Vaudry
- From the ‡Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, 76000 Rouen, France;
- §Normandie Univ, UNIROUEN, Rouen Proteomic Platform (PISSARO), Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
- ¶Normandie Univ, UNIROUEN, Regional Cell Imaging Platform of Normandy (PRIMACEN), 76000 Rouen, France
| |
Collapse
|
27
|
Abstract
This paper is the thirty-ninth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2016 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and CUNY Neuroscience Collaborative, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
28
|
Zaveri NT, Marquez PV, Meyer ME, Polgar WE, Hamid A, Lutfy K. A Novel and Selective Nociceptin Receptor (NOP) Agonist (1-(1-((cis)-4-isopropylcyclohexyl)piperidin-4-yl)-1H-indol-2-yl)methanol (AT-312) Decreases Acquisition of Ethanol-Induced Conditioned Place Preference in Mice. Alcohol Clin Exp Res 2018; 42:461-471. [PMID: 29215139 DOI: 10.1111/acer.13575] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 11/30/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Nociceptin/orphanin FQ, the endogenous peptide agonist for the opioid receptor-like receptor (also known as NOP or the nociceptin receptor), has been shown to block the acquisition and expression of ethanol (EtOH)-induced conditioned place preference (CPP). Here, we report the characterization of a novel small-molecule NOP ligand AT-312 (1-(1-((cis)-4-isopropylcyclohexyl)piperidin-4-yl)-1H-indol-2-yl)methanol) in receptor binding and GTPγS functional assays in vitro. We then investigated the effect of AT-312 on the rewarding action of EtOH in mice using the CPP paradigm. Further, using mice lacking the NOP receptor and their wild-type controls, we also examined the involvement of NOP in the effect of AT-312. Motivational effects of AT-312 alone were also assessed in the CPP paradigm. METHODS Female mice lacking NOP and/or their wild-type controls received conditioning in the presence or absence of the NOP agonist [AT-312 (1, 3, and 10 mg/kg) or the control NOP agonist SCH221510 (10 mg/kg)] followed by saline/EtOH for 3 consecutive days (twice daily) and tested for CPP in a drug-free state on the next day. RESULTS Our in vitro data showed that AT-312 is a high-affinity, selective NOP full agonist with 17-fold selectivity over the mu opioid receptor and >200-fold selectivity over the kappa opioid receptor. The results of our in vivo studies showed that AT-312 reduced EtOH CPP at the lowest dose (1 mg/kg) tested but completely abolished EtOH CPP at higher doses (3 or 10 mg/kg) compared to their vehicle-treated control group. AT-312 (3 mg/kg) did not alter EtOH-induced CPP in mice lacking NOP, confirming that AT-312 reduced EtOH CPP through its action at the NOP receptor. AT-312 (3 mg/kg) did not induce reward or aversion when administered alone, showing that the novel small-molecule NOP agonist shows efficacy in blocking EtOH-induced CPP via the NOP receptor. CONCLUSIONS Together, these data suggest that small-molecule NOP agonists have the potential to reduce alcohol reward and may be promising as medications to treat alcohol addiction.
Collapse
Affiliation(s)
| | - Paul V Marquez
- College of Pharmacy, Western University of Health Sciences, Pomona, California
| | | | | | - Abdul Hamid
- College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Kabirullah Lutfy
- College of Pharmacy, Western University of Health Sciences, Pomona, California
| |
Collapse
|
29
|
Della Longa S, Arcovito A. “In silico” study of the binding of two novel antagonists to the nociceptin receptor. J Comput Aided Mol Des 2018; 32:385-400. [DOI: 10.1007/s10822-017-0095-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/29/2017] [Indexed: 01/25/2023]
|
30
|
Novelle MG, Diéguez C. Food Addiction and Binge Eating: Lessons Learned from Animal Models. Nutrients 2018; 10:E71. [PMID: 29324652 PMCID: PMC5793299 DOI: 10.3390/nu10010071] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 12/26/2017] [Accepted: 01/09/2018] [Indexed: 01/10/2023] Open
Abstract
The feeding process is required for basic life, influenced by environment cues and tightly regulated according to demands of the internal milieu by regulatory brain circuits. Although eating behaviour cannot be considered "addictive" under normal circumstances, people can become "addicted" to this behaviour, similarly to how some people are addicted to drugs. The symptoms, cravings and causes of "eating addiction" are remarkably similar to those experienced by drug addicts, and both drug-seeking behaviour as eating addiction share the same neural pathways. However, while the drug addiction process has been highly characterised, eating addiction is a nascent field. In fact, there is still a great controversy over the concept of "food addiction". This review aims to summarize the most relevant animal models of "eating addictive behaviour", emphasising binge eating disorder, that could help us to understand the neurobiological mechanisms hidden under this behaviour, and to improve the psychotherapy and pharmacological treatment in patients suffering from these pathologies.
Collapse
Affiliation(s)
- Marta G Novelle
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria (IDIS), CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 15786 Santiago de Compostela, Spain.
| | - Carlos Diéguez
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria (IDIS), CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 15786 Santiago de Compostela, Spain.
| |
Collapse
|
31
|
Genovese RF, Dobre S. Mitigation of adverse behavioral impact from predator exposure by the nociceptin/orphanin FQ peptide antagonist J-113397 in rats. Behav Pharmacol 2017; 28:521-530. [PMID: 28704271 DOI: 10.1097/fbp.0000000000000329] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The nociceptin/orphanin FQ peptide (NOP) receptor is believed to have an integral modulatory function in the stress response system. We evaluated the highly selective NOP antagonist J-113397 (7.5 and 20.0 mg/kg), using a predator exposure in which rats were exposed to predator cats as a stressor. A single dose of J-113397 or vehicle was administered (intraperitoneally) shortly before exposure to the predators or a sham exposure. Behavioral impact was measured using elevated plus maze (EPM), open field activity (OFA), and an olfactory discrimination (OD). The predator exposure produced a relatively long-lasting deficit (decreased time in open arms, decreased basic activity) on the EPM while having little effect on performance on the OFA or OD. J-113397 mitigated the performance deficits on the EPM in a dose-dependent manner while having little effect on performance on the OFA or OD. The largest dose of J-113397, administered with a sham exposure, was essentially devoid of effects on the EPM, OFA, and OD. These results demonstrate that J-113397 can significantly and selectively mitigate the effects of a stressor typically used in a preclinical model of post-traumatic stress disorder. Furthermore, these results are consistent with and extend previous results showing that the NOP receptor has an important role in the response to stress and that NOP antagonism may, potentially, have therapeutic benefit in stress disorders.
Collapse
Affiliation(s)
- Raymond F Genovese
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | |
Collapse
|
32
|
Abstract
Binge-eating disorder (BED) is the most prevalent eating disorder with estimates of 2-5% of the general adult population. Nonetheless, its pathophysiology is poorly understood. Furthermore, there exist few therapeutic options for its effective treatment. Here we review the current state of binge-eating neurobiology and pharmacology, drawing from clinical therapeutic, neuroimaging, cognitive, human genetic and animal model studies. These studies, which are still in their infancy, indicate that while there are many gaps in our knowledge, several key neural substrates appear to underpin binge-eating and may be conserved between human and animals. This observation suggests that behavioral intermediate phenotypes or endophenotypes relevant to BED may be modeled in animals, facilitating the identification and testing of novel pharmacological targets. The development of novel, safe and effective pharmacological therapies for the treatment of BED will enhance the ability of clinicians to provide optimal care for people with BED.
Collapse
Affiliation(s)
- Peter H Hutson
- Department of Neurobiology, CNS Discovery, Teva Pharmaceuticals, West Chester, PA, USA.
| | - Iris M Balodis
- Peter Boris Centre for Addiction Research, Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Marc N Potenza
- Department of Psychiatry, Child Study Center, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA; National Center on Addiction and Substance Abuse, USA; Connecticut Mental Health Center, New Haven, CT, USA
| |
Collapse
|
33
|
Abstract
Eating disorders (EDs), including anorexia nervosa, bulimia nervosa, and binge-eating disorder, constitute a class of common and deadly psychiatric disorders. While numerous studies in humans highlight the important role of neurobiological alterations in the development of ED-related behaviors, the precise neural substrate that mediates this risk is unknown. Historically, pharmacological interventions have played a limited role in the treatment of eating disorders, typically providing symptomatic relief of comorbid psychiatric issues, like depression and anxiety, in support of the standard nutritional and psychological treatments. To date there are no Food and Drug Administration-approved medications or procedures for anorexia nervosa, and only one Food and Drug Administration-approved medication each for bulimia nervosa (fluoxetine) and binge-eating disorder (lisdexamfetamine). While there is little primary interest in drug development for eating disorders, postmarket monitoring of medications and procedures approved for other indications has identified several novel treatment options for patients with eating disorders. In this review, I utilize searches of the PubMed and ClinicalTrials.gov databases to highlight emerging treatments in eating disorders.
Collapse
Affiliation(s)
- Michael Lutter
- Eating Recovery Center of Dallas, 4716 Alliance Blvd. #400, Plano, TX, 75093, USA.
| |
Collapse
|
34
|
Abstract
This paper is the thirty-eighth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2015 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
35
|
Kallupi M, Scuppa G, de Guglielmo G, Calò G, Weiss F, Statnick MA, Rorick-Kehn LM, Ciccocioppo R. Genetic Deletion of the Nociceptin/Orphanin FQ Receptor in the Rat Confers Resilience to the Development of Drug Addiction. Neuropsychopharmacology 2017; 42:695-706. [PMID: 27562376 PMCID: PMC5240182 DOI: 10.1038/npp.2016.171] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 12/14/2022]
Abstract
The nociceptin (NOP) receptor is a G-protein-coupled receptor whose natural ligand is the NOP/orphanin FQ (N/OFQ) peptide. Evidence from pharmacological studies suggests that the N/OFQ system is implicated in the regulation of several addiction-related phenomena, such as drug intake, withdrawal, and relapse. Here, to further explore the role of NOP system in addiction, we used NOP (-/-) rats to study the motivation for cocaine, heroin, and alcohol self-administration in the absence of N/OFQ function. Conditioned place preference (CPP) and saccharin (0.2% w/v) self-administration were also investigated. Results showed that NOP (-/-) rats self-administer less cocaine (0.25, 0.125, or 0.5 mg/infusion) both under a fixed ratio 1 and a progressive ratio schedule of reinforcement compared with wild-type (Wt) controls. Consistently, cocaine (10 mg/kg, i.p.) was able to induce CPP in Wt but not in NOP (-/-). When NOP (-/-) rats were tested for heroin (20 μg/infusion) and ethanol (10% v/v) self-administration, they showed significantly lower drug intake compared with Wt. Conversely, saccharin self-administration was not affected by NOP deletion, excluding the possibility of nonspecific learning deficits or generalized disruption of reward mechanisms in NOP (-/-) rats. These findings were confirmed with pharmacological experiments using two selective NOP antagonists, SB-612111 and LY2817412. Both drugs attenuated alcohol self-administration in Wt rats but not in NOP (-/-) rats. In conclusion, our results demonstrate that genetic deletion of NOP receptors confers resilience to drug abuse and support a role for NOP receptor antagonism as a potential treatment option for drug addiction.
Collapse
Affiliation(s)
- Marsida Kallupi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Giulia Scuppa
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Giordano de Guglielmo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Girolamo Calò
- Department of Medical Science, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Friedbert Weiss
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA
| | - Michael A Statnick
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN USA
| | | | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032 Italy, Tel: +39 07 3740 3313, Fax: +39 07 3740 3325, E-mail:
| |
Collapse
|
36
|
DEMİR ÖZKAY Ü, CAN ÖD, TURAN N, ÇAVUŞOĞLU KAYA B. Synthesis and antinociceptive activities of some novel benzimidazole-piperidine derivatives. Turk J Chem 2017. [DOI: 10.3906/kim-1612-76] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
37
|
Witkin JM, Rorick-Kehn LM, Benvenga MJ, Adams BL, Gleason SD, Knitowski KM, Li X, Chaney S, Falcone JF, Smith JW, Foss J, Lloyd K, Catlow JT, McKinzie DL, Svensson KA, Barth VN, Toledo MA, Diaz N, Lafuente C, Jiménez A, Benito A, Pedregal C, Martínez-Grau MA, Post A, Ansonoff MA, Pintar JE, Statnick MA. Preclinical findings predicting efficacy and side-effect profile of LY2940094, an antagonist of nociceptin receptors. Pharmacol Res Perspect 2016; 4:e00275. [PMID: 28097008 PMCID: PMC5226289 DOI: 10.1002/prp2.275] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/05/2016] [Accepted: 10/12/2016] [Indexed: 12/28/2022] Open
Abstract
Nociceptin/Orphanin FQ (N/OFQ) is a 17 amino acid peptide whose receptor is designated ORL1 or nociceptin receptor (NOP). We utilized a potent, selective, and orally bioavailable antagonist with documented engagement with NOP receptors in vivo to assess antidepressant‐ and anxiolytic‐related pharmacological effects of NOP receptor blockade along with measures of cognitive and motor impingement. LY2940094 ([2‐[4‐[(2‐chloro‐4,4‐difluoro‐spiro[5H‐thieno[2,3‐c]pyran‐7,4′‐piperidine]‐1′‐yl)methyl]‐3‐methyl‐pyrazol‐1‐yl]‐3‐pyridyl]methanol) displayed antidepressant‐like behavioral effects in the forced‐swim test in mice, an effect absent in NOP−/− mice. LY2940094 also augmented the behavioral effect of fluoxetine without changing target occupancies (NOP and serotonin reuptake transporter [SERT]). LY2940094 did not have effects under a differential‐reinforcement of low rate schedule. Although anxiolytic‐like effects were not observed in some animal models (conditioned suppression, 4‐plate test, novelty‐suppressed feeding), LY2940094 had effects like that of anxiolytic drugs in three assays: fear‐conditioned freezing in mice, stress‐induced increases in cerebellar cGMP in mice, and stress‐induced hyperthermia in rats. These are the first reports of anxiolytic‐like activity with a systemically viable NOP receptor antagonist. LY2940094 did not disrupt performance in either a 5‐choice serial reaction time or delayed matching‐to‐position assay. LY2940094 was also not an activator or suppressor of locomotion in rodents nor did it induce failures of rotarod performance. These data suggest that LY2940094 has unique antidepressant‐ and anxiolytic‐related pharmacological effects in rodents. Clinical proof of concept data on this molecule in depressed patients have been reported elsewhere.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | | | - Mark J Benvenga
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Benjamin L Adams
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Scott D Gleason
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Karen M Knitowski
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Xia Li
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Steven Chaney
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Julie F Falcone
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Janice W Smith
- Lilly Research Laboratories Eli Lilly and Company Windlesham Surrey United Kingdom
| | - Julie Foss
- Lilly Research Laboratories Eli Lilly and Company Windlesham Surrey United Kingdom
| | - Kirsti Lloyd
- Lilly Research Laboratories Eli Lilly and Company Windlesham Surrey United Kingdom
| | - John T Catlow
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - David L McKinzie
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Kjell A Svensson
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Vanessa N Barth
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana
| | - Miguel A Toledo
- Lilly Research Laboratories Eli Lilly and Company Indianapolis Indiana; Lilly Research Laboratories Eli Lilly and Company Alcobendas Madrid Spain
| | - Nuria Diaz
- Lilly Research Laboratories Eli Lilly and Company Alcobendas Madrid Spain
| | - Celia Lafuente
- Lilly Research Laboratories Eli Lilly and Company Alcobendas Madrid Spain
| | - Alma Jiménez
- Lilly Research Laboratories Eli Lilly and Company Alcobendas Madrid Spain
| | - Alfonso Benito
- Lilly Research Laboratories Eli Lilly and Company Alcobendas Madrid Spain
| | | | | | - Anke Post
- Lilly Research Laboratories Eli Lilly and Company Windlesham Surrey United Kingdom
| | - Michael A Ansonoff
- Lilly Research Laboratories Eli Lilly and Company Rutgers-Robert Wood Johnson Medical School New Brunswick New Jersey
| | - John E Pintar
- Lilly Research Laboratories Eli Lilly and Company Rutgers-Robert Wood Johnson Medical School New Brunswick New Jersey
| | | |
Collapse
|
38
|
Gaetani S, Romano A, Provensi G, Ricca V, Lutz T, Passani MB. Eating disorders: from bench to bedside and back. J Neurochem 2016; 139:691-699. [DOI: 10.1111/jnc.13848] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Silvana Gaetani
- Department of Physiology and Pharmacology “V. Erspamer”; Sapienza University of Rome; Rome Italy
| | - Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer”; Sapienza University of Rome; Rome Italy
| | - Gustavo Provensi
- Department of Neuroscience, Psychology, Drug Discovery and Child Health (NEUROFARBA); University of Florence; Florence Italy
| | - Valdo Ricca
- Department of Neuroscience, Psychology, Drug Discovery and Child Health (NEUROFARBA); University of Florence; Florence Italy
| | - Thomas Lutz
- Institute of Veterinary Physiology; Vetsuisse Faculty University of Zurich; Zurich Switzerland
- Center of Integrative Human Physiology; University of Zurich; Zurich Switzerland
| | | |
Collapse
|
39
|
Mendez IA, Maidment NT, Murphy NP. Parsing the hedonic and motivational influences of nociceptin on feeding using licking microstructure analysis in mice. Behav Pharmacol 2016; 27:516-27. [PMID: 27100061 PMCID: PMC4965319 DOI: 10.1097/fbp.0000000000000240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Opioid peptides are implicated in processes related to reward and aversion; however, how specific opioid peptides are involved remains unclear. We investigated the role of nociceptin (NOC) in voluntary licking for palatable and aversive tastants by studying the effect of intracerebroventricularly administered NOC on licking microstructure in wild-type and NOC receptor knockout (NOP KO) mice. Compared with the wild-type mice, NOP KO mice emitted fewer bouts of licking when training to lick for a 20% sucrose solution. Correspondingly, intracerebroventricular administration of NOC increased the number of licking bouts for sucrose and sucralose in wild-type, but not in NOP KO mice. The ability of NOC to initiate new bouts of licking for sweet solutions suggests that NOC may drive motivational aspects of feeding behavior. Conversely, adulterating a sucrose solution with the aversive tastant quinine reduced licking bout lengths in wild-type and NOP KOs, suggesting that NOC signaling is not involved in driving voluntary consumption of semiaversive tastants. Interestingly, when consuming sucrose following 20 h of food deprivation, NOP KO mice emitted longer bouts of licking than wild types, suggesting that under hungry conditions, NOC may also contribute toward hedonic aspects of feeding. Together, these results suggest differential roles for NOC in the motivational and hedonic aspects of feeding.
Collapse
Affiliation(s)
- Ian A Mendez
- Department of Psychiatry and Biobehavioral Sciences, Hatos Center, Semel Institute for Neuroscience and Human Behavior, University of California at Los Angeles, Los Angeles, California, USA
| | | | | |
Collapse
|
40
|
Post A, Smart TS, Jackson K, Mann J, Mohs R, Rorick-Kehn L, Statnick M, Anton R, O'Malley SS, Wong CJ. Proof-of-Concept Study to Assess the Nociceptin Receptor Antagonist LY2940094 as a New Treatment for Alcohol Dependence. Alcohol Clin Exp Res 2016; 40:1935-44. [DOI: 10.1111/acer.13147] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 06/03/2016] [Indexed: 12/13/2022]
Affiliation(s)
| | | | | | | | | | | | | | - Raymond Anton
- Medical University of South Carolina; Charleston South Carolina
| | | | | |
Collapse
|
41
|
Hardaway JA, Jensen J, Kim M, Mazzone CM, Sugam JA, Diberto JF, Lowery-Gionta EG, Hwa LS, Pleil KE, Bulik CM, Kash TL. Nociceptin receptor antagonist SB 612111 decreases high fat diet binge eating. Behav Brain Res 2016; 307:25-34. [PMID: 27036650 PMCID: PMC4896639 DOI: 10.1016/j.bbr.2016.03.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/25/2016] [Accepted: 03/28/2016] [Indexed: 10/22/2022]
Abstract
Binge eating is a dysregulated form of feeding behavior that occurs in multiple eating disorders including binge-eating disorder, the most common eating disorder. Feeding is a complex behavioral program supported through the function of multiple brain regions and influenced by a diverse array of receptor signaling pathways. Previous studies have shown the overexpression of the opioid neuropeptide nociceptin (orphanin FQ, N/OFQ) can induce hyperphagia, but the role of endogenous nociceptin receptor (NOP) in naturally occurring palatability-induced hyperphagia is unknown. In this study we adapted a simple, replicable form of binge eating of high fat food (HFD). We found that male and female C57BL/6J mice provided with daily one-hour access sessions to HFD eat significantly more during this period than those provided with continuous 24h access. This form of feeding is rapid and entrained. Chronic intermittent HFD binge eating produced hyperactivity and increased light zone exploration in the open field and light-dark assays respectively. Treatment with the potent and selective NOP antagonist SB 612111 resulted in a significant dose-dependent reduction in binge intake in both male and female mice, and, unlike treatment with the serotonin selective reuptake inhibitor fluoxetine, produced no change in total 24-h food intake. SB 612111 treatment also significantly decreased non-binge-like acute HFD consumption in male mice. These data are consistent with the hypothesis that high fat binge eating is modulated by NOP signaling and that the NOP system may represent a promising novel receptor to explore for the treatment of binge eating.
Collapse
Affiliation(s)
- J Andrew Hardaway
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Jennifer Jensen
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Michelle Kim
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Christopher M Mazzone
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Jonathan A Sugam
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Jeffrey F Diberto
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Emily G Lowery-Gionta
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Lara S Hwa
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Kristen E Pleil
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA
| | - Cynthia M Bulik
- UNC Department of Psychiatry, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Nutrition, University of North Carolina at Chapel Hill, NC, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, NC, USA; UNC Department of Pharmacology, University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
42
|
Raddad E, Chappell A, Meyer J, Wilson A, Ruegg CE, Tauscher J, Statnick MA, Barth V, Zhang X, Verfaille SJ. Occupancy of Nociceptin/Orphanin FQ Peptide Receptors by the Antagonist LY2940094 in Rats and Healthy Human Subjects. Drug Metab Dispos 2016; 44:1536-42. [DOI: 10.1124/dmd.116.070359] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/06/2016] [Indexed: 11/22/2022] Open
|
43
|
Rorick-Kehn LM, Ciccocioppo R, Wong CJ, Witkin JM, Martinez-Grau MA, Stopponi S, Adams BL, Katner JS, Perry KW, Toledo MA, Diaz N, Lafuente C, Jiménez A, Benito A, Pedregal C, Weiss F, Statnick MA. A Novel, Orally Bioavailable Nociceptin Receptor Antagonist, LY2940094, Reduces Ethanol Self-Administration and Ethanol Seeking in Animal Models. Alcohol Clin Exp Res 2016; 40:945-54. [PMID: 27084498 DOI: 10.1111/acer.13052] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/24/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND The nociceptin/orphanin-FQ (or opioid receptor-like [ORL1]) receptor (NOP) is localized in the mesolimbic reward pathway and has been suggested to play a role in feeding, mood, stress, and addiction. Since its deorphanization in 1995, there has been a clear dichotomy in the literature regarding whether an agonist or antagonist would provide therapeutic benefit. Specifically, the literature reports indicate that NOP receptor antagonists produce efficacy in animal models of hyperphagia and antidepressant-like activity, whereas NOP agonists produce anxiolytic-like effects and dampen reward/addiction behaviors including ethanol consumption. METHODS We characterize here the potent, orally bioavailable NOP antagonist, LY2940094, in rodent models of ethanol consumption, including ethanol self-administration, progressive ratio operant self-administration, stress-induced reinstatement of ethanol seeking, and in vivo microdialysis in the nucleus accumbens. RESULTS LY2940094 dose dependently reduced homecage ethanol self-administration in Indiana alcohol-preferring (P) and Marchigian Sardinian alcohol-preferring (msP) rats, without affecting food/water intake or locomotor activity. Reduced ethanol intake in P rats did not show significant tolerance over 4 days of subchronic dosing. LY2940094 attenuated progressive ratio operant responding and break points for ethanol in P rats. Moreover, stress-induced reinstatement of ethanol seeking in msP rats was completely blocked by LY2940094. Furthermore, LY2940094 blocked ethanol-stimulated dopamine release in response to ethanol challenge (1.1 g/kg, intraperitoneally). CONCLUSIONS Our findings demonstrate for the first time that blockade of NOP receptors attenuates ethanol self-administration and ethanol-motivated behaviors, stress-induced ethanol seeking, and ethanol-induced stimulation of brain reward pathways in lines of rats that exhibit excessive ethanol consumption. Results suggest that LY2940094 may have potential therapeutic utility in treating alcohol addiction.
Collapse
Affiliation(s)
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Conrad J Wong
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Jeffrey M Witkin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | | | - Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Benjamin L Adams
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Jason S Katner
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Kenneth W Perry
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | | | | | - Friedbert Weiss
- Department of Molecular and Cellular Neurosciences, The Scripps Research Institute, La Jolla, California
| | - Michael A Statnick
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| |
Collapse
|