1
|
Staedtler ES, Sapio MR, King DM, Maric D, Ghetti A, Mannes AJ, Iadarola MJ. The μ-opioid receptor differentiates two distinct human nociceptive populations relevant to clinical pain. Cell Rep Med 2024; 5:101788. [PMID: 39413733 PMCID: PMC11513826 DOI: 10.1016/j.xcrm.2024.101788] [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: 11/27/2023] [Revised: 06/26/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
The shortfall in new analgesic agents is a major impediment to reducing reliance on opioid medications for control of severe pain. In both animals and man, attenuating nociceptive transmission from primary afferent neurons with a μ-opioid receptor agonist yields highly effective analgesia. Consequently, deeper molecular characterization of human nociceptive afferents expressing OPRM1, the μ-opioid receptor gene, is a key component for advancing analgesic drug discovery and understanding clinical pain control. A co-expression matrix for the μ-opioid receptor and a variety of nociceptive channels as well as δ- and κ-opioid receptors is established by multiplex in situ hybridization. Our results indicate an OPRM1-positive population with strong molecular resemblance to rodent peptidergic C-nociceptors associated with tissue damage pain and an OPRM1-negative population sharing molecular characteristics of murine non-peptidergic C-nociceptors. The empirical identification of two distinct human nociceptive populations that differ profoundly in their presumed responsiveness to opioids provides an actionable translational framework for human pain control.
Collapse
Affiliation(s)
- Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | | | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Sayers S, Le N, Hernandez J, Mata‐Pacheco V, Wagner EJ. The vital role of arcuate nociceptin/orphanin FQ neurones in mounting an oestradiol-dependent adaptive response to negative energy balance via inhibition of nearby proopiomelanocortin neurones. J Physiol 2022; 600:4939-4961. [PMID: 36217719 PMCID: PMC9828807 DOI: 10.1113/jp283378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023] Open
Abstract
We tested the hypothesis that N/OFQ neurones in the arcuate nucleus (N/OFQARC ) inhibit proopiomelanocortin (POMCARC ) neurones in a diet- and hormone-dependent manner to promote a more extensive rebound hyperphagia upon re-feeding following an 18 h fast. We utilized intact male or ovariectomized (OVX) female mice subjected to ad libitum-feeding or fasting conditions. N/OFQARC neurones under negative energy balance conditions displayed heightened sensitivity as evidenced by a decreased rheobase threshold, increased firing frequency, and increased burst duration and frequency compared to ad libitum-feeding conditions. Stimulation of N/OFQARC neurones more robustly inhibited POMCARC neurones under fasting conditions compared to ad libitum-feeding conditions. N/OFQARC inhibition of POMCARC neurones is hormone dependent as chemostimulation of N/OFQARC neurones from fasted males and OVX females produced a sizable outward current in POMCARC neurones. Oestradiol (E2 ) markedly attenuated the N/OFQ-induced POMCARC outward current. Additionally, N/OFQ tonically inhibits POMCARC neurones to a greater degree under fasting conditions than in ad libitum-feeding conditions as evidenced by the abrogation of N/OFQ-nociceptin opioid peptide (NOP) receptor signalling and inhibition of N/OFQ release via chemoinhibition of N/OFQARC neurones. Intra-arcuate nucleus application of N/OFQ further elevated the hyperphagic response and increased meal size during the 6 h re-feed period, and these effects were mimicked by chemostimulation of N/OFQARC neurones in vivo. E2 attenuated the robust N/OFQ-induced rebound hyperphagia seen in vehicle-treated OVX females. These data demonstrate that N/OFQARC neurones play a vital role in mitigating the impact of negative energy balance by inhibiting the excitability of anorexigenic neural substrates, an effect that is diminished by E2 in females. KEY POINTS: Nociceptin/orphanin FQ (N/OFQ) promotes increased energy intake and decreased energy expenditure under conditions of positive energy balance in a sex- and hormone-dependent manner. Here it is shown that under conditions of negative energy balance, i.e. fasting, N/OFQ inhibits anorexigenic proopiomelanocortin (POMC) neurones to a greater degree compared to homeostatic conditions due to fasting-induced hyperexcitability of N/OFQ neurones. Additionally, N/OFQ promotes a sustained increase in rebound hyperphagia and increase in meal size during the re-feed period following a fast. These results promote greater understanding of how energy balance influences the anorexigenic circuitry of the hypothalamus, and aid in understanding the neurophysiological pathways implicated in eating disorders promoting cachexia.
Collapse
Affiliation(s)
- Sarah Sayers
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Nikki Le
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Jennifer Hernandez
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Veronica Mata‐Pacheco
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Edward J. Wagner
- College of Osteopathic Medicine of the PacificWestern University of Health SciencesPomonaCAUSA
| |
Collapse
|
3
|
Dworsky-Fried Z, Chadwick CI, Kerr BJ, Taylor AMW. Multiple Sclerosis and the Endogenous Opioid System. Front Neurosci 2021; 15:741503. [PMID: 34602975 PMCID: PMC8484329 DOI: 10.3389/fnins.2021.741503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by chronic inflammation, neuronal degeneration and demyelinating lesions within the central nervous system. The mechanisms that underlie the pathogenesis and progression of MS are not fully known and current therapies have limited efficacy. Preclinical investigations using the murine experimental autoimmune encephalomyelitis (EAE) model of MS, as well as clinical observations in patients with MS, provide converging lines of evidence implicating the endogenous opioid system in the pathogenesis of this disease. In recent years, it has become increasingly clear that endogenous opioid peptides, binding μ- (MOR), κ- (KOR) and δ-opioid receptors (DOR), function as immunomodulatory molecules within both the immune and nervous systems. The endogenous opioid system is also well known to play a role in the development of chronic pain and negative affect, both of which are common comorbidities in MS. As such, dysregulation of the opioid system may be a mechanism that contributes to the pathogenesis of MS and associated symptoms. Here, we review the evidence for a connection between the endogenous opioid system and MS. We further explore the mechanisms by which opioidergic signaling might contribute to the pathophysiology and symptomatology of MS.
Collapse
Affiliation(s)
- Zoë Dworsky-Fried
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Caylin I. Chadwick
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Bradley J. Kerr
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, AB, Canada
| | - Anna M. W. Taylor
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
4
|
Schaffer A, Hajagos-Tóth J, Ducza E, Bódi N, Bagyánszki M, Szalai Z, Gáspár R. The ontogeny of kisspeptin receptor in the uterine contractions in rats: Its possible role in the quiescence of non-pregnant and pregnant uteri. Eur J Pharmacol 2021; 896:173924. [PMID: 33548216 DOI: 10.1016/j.ejphar.2021.173924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/20/2021] [Accepted: 01/29/2021] [Indexed: 10/22/2022]
Abstract
The objectives of this study were to investigate the effects of KISS1 94-121 fragment on the contractility of non-pregnant and pregnant rat uteri, and to determine the uterine and myometrial expressions of Kiss1r. Uterine muscle strips were obtained from non-pregnant Sprague-Dawley rats in oestrous phase and from pregnant rats on gestational days 5, 15, 18, 20 or 22. The in vitro contractility measurements were carried out in an isolated organ bath in the presence of KISS1 94-121. Experiments with 5-day pregnant tissues were also performed in the presence of kisspeptin-234 trifluoroacetate. The mRNA and protein expressions of Kiss1r were measured by RT-PCR and Western blot analysis, while localizations of receptors were defined by fluorescent immunohistochemistry. KISS1 94-121 induced a dose-dependent relaxation both in non-pregnant and pregnant intact and endometrium-denuded uteri. A gradual decrease was found in the uterine expressions of Kiss1r mRNA and protein towards the end of the gestational period, and it was further confirmed by the immunohistochemical results. The significant majority of Kiss1r is found in the myometrium, however the few endometrial Kiss1r also influences the uterine contractions. The relaxing effect of kisspeptin is continuously reduced towards the end of gestational period in parallel with the reduction of Kiss1r expression. Our results suggest a putative role of kisspeptin in the maintenance of uterine quiescence that may have significance in miscarriage or preterm contractions.
Collapse
Affiliation(s)
- Annamária Schaffer
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Hungary
| | - Judit Hajagos-Tóth
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Hungary
| | - Eszter Ducza
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Hungary
| | - Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, Institute of Biology, Faculty of Science and Informatics, University of Szeged, Hungary
| | - Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, Institute of Biology, Faculty of Science and Informatics, University of Szeged, Hungary
| | - Zita Szalai
- Department of Physiology, Anatomy and Neuroscience, Institute of Biology, Faculty of Science and Informatics, University of Szeged, Hungary
| | - Róbert Gáspár
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Hungary.
| |
Collapse
|
5
|
Abstract
Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug-seeking and drug-taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central nucleus of the amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptive mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid (GABA)ergic transmission in the CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the CeA, whereas chronic alcohol up-regulates NMDA receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and antistress (e.g., nociceptin/orphanin FQ, oxytocin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.
Collapse
Affiliation(s)
- Marisa Roberto
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Dean Kirson
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Sophia Khom
- Departments of Molecular Medicine and Neuroscience, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
6
|
Targowska-Duda KM, Ozawa A, Bertels Z, Cippitelli A, Marcus JL, Mielke-Maday HK, Zribi G, Rainey AN, Kieffer BL, Pradhan AA, Toll L. NOP receptor agonist attenuates nitroglycerin-induced migraine-like symptoms in mice. Neuropharmacology 2020; 170:108029. [PMID: 32278976 PMCID: PMC7243257 DOI: 10.1016/j.neuropharm.2020.108029] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 01/02/2023]
Abstract
Migraine is an extraordinarily prevalent and disabling headache disorder that affects one billion people worldwide. Throbbing pain is one of several migraine symptoms including sensitivity to light (photophobia), sometimes to sounds, smell and touch. The basic mechanisms underlying migraine remain inadequately understood, and current treatments (with triptans being the primary standard of care) are not well tolerated by some patients. NOP (Nociceptin OPioid) receptors, the fourth member of the opioid receptor family, are expressed in the brain and periphery with particularly high expression known to be in trigeminal ganglia (TG). The aim of our study was to further explore the involvement of the NOP receptor system in migraine. To this end, we used immunohistochemistry to examine NOP receptor distribution in TG and trigeminal nucleus caudalus (TNC) in mice, including colocalization with specific cellular markers, and used nitroglycerin (NTG) models of migraine to assess the influence of the selective NOP receptor agonist, Ro 64-6198, on NTG-induced pain (sensitivity of paw and head using von Frey filaments) and photophobia in mice. Our immunohistochemical studies with NOP-eGFP knock-in mice indicate that NOP receptors are on the majority of neurons in the TG and are also very highly expressed in the TNC. In addition, Ro 64-6198 can dose dependently block NTG-induced paw and head allodynia, an effect that is blocked by the NOP antagonist, SB-612111. Moreover, Ro 64-6198, can decrease NTG-induced light sensitivity in mice. These results suggest that NOP receptor agonists should be futher explored as treatment for migraine symptoms. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Katarzyna M Targowska-Duda
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, United States; Department of Biopharmacy, Medical University of Lublin, Lublin, Poland
| | - Akihiko Ozawa
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Zachariah Bertels
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Andrea Cippitelli
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Jason L Marcus
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Hanna K Mielke-Maday
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Gilles Zribi
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Amanda N Rainey
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Brigitte L Kieffer
- Douglas Hospital Research Center, Dep. of Psychiatry, School of Medicine, McGill University, Montreal, Quebec, Canada; INSERM U1114, Strasbourg, France
| | - Amynah A Pradhan
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Lawrence Toll
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, United States.
| |
Collapse
|
7
|
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: 24] [Impact Index Per Article: 4.8] [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
|
8
|
Rogers TJ. Bidirectional Regulation of Opioid and Chemokine Function. Front Immunol 2020; 11:94. [PMID: 32076421 PMCID: PMC7006827 DOI: 10.3389/fimmu.2020.00094] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
The opioid family of GPCRs consists of the classical opioid receptors, designated μ-, κ-, and δ-opioid receptors, and the orphanin-FQ receptor, and these proteins are expressed on both neuronal and hematopoietic cells. A number of laboratories have reported that an important degree of cross-talk can occur between the opioid receptors and the chemokine and chemokine receptor families. As a part of this, the opioid receptors are known to regulate the expression of certain chemokines and chemokine receptors, including those that possess strong pro-inflammatory activity. At the level of receptor function, it is clear that certain members of the chemokine family can mediate cross-desensitization of the opioid receptors. Conversely, the opioid receptors are all able to induce heterologous desensitization of some of the chemokine receptors. Consequently, activation of one or more of the opioid receptors can selectively cross-desensitize chemokine receptors and regulate chemokine function. These cross-talk processes have significant implications for the inflammatory response, since the regulation of both the recruitment of inflammatory cells, as well as the sensation of pain, can be controlled in this way.
Collapse
Affiliation(s)
- Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
9
|
Torres-Berrio A, Nava-Mesa MO. The opioid system in stress-induced memory disorders: From basic mechanisms to clinical implications in post-traumatic stress disorder and Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:327-338. [PMID: 30118823 DOI: 10.1016/j.pnpbp.2018.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/25/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
Abstract
Cognitive and emotional impairment are a serious consequence of stress exposure and are core features of neurological and psychiatric conditions that involve memory disorders. Indeed, acute and chronic stress are high-risk factors for the onset of post-traumatic stress disorder (PTSD) and Alzheimer's disease (AD), two devastating brain disorders associated with memory dysfunction. Besides the sympathetic nervous system and the hypothalamic-pituitary-adrenal (HPA) axis, stress response also involves the activation of the opioid system in brain regions associated with stress regulation and memory processing. In this context, it is possible that stress-induced memory disorders may be attributed to alterations in the interaction between the neuroendocrine stress system and the opioid system. In this review, we: (1) describe the effects of acute and chronic stress on memory, and the modulatory role of the opioid system, (2) discuss the contribution of the opioid system to the pathophysiology of PTSD and AD, and (3) present evidence of current and potential therapies that target the opioid receptors to treat PTSD- and AD-associated symptoms.
Collapse
Affiliation(s)
| | - Mauricio O Nava-Mesa
- Neuroscience Research Group (NEUROS), School of Medicine, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
10
|
Lippmann C, Kringel D, Ultsch A, Lötsch J. Computational functional genomics-based approaches in analgesic drug discovery and repurposing. Pharmacogenomics 2018; 19:783-797. [DOI: 10.2217/pgs-2018-0036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Persistent pain is a major healthcare problem affecting a fifth of adults worldwide with still limited treatment options. The search for new analgesics increasingly includes the novel research area of functional genomics, which combines data derived from various processes related to DNA sequence, gene expression or protein function and uses advanced methods of data mining and knowledge discovery with the goal of understanding the relationship between the genome and the phenotype. Its use in drug discovery and repurposing for analgesic indications has so far been performed using knowledge discovery in gene function and drug target-related databases; next-generation sequencing; and functional proteomics-based approaches. Here, we discuss recent efforts in functional genomics-based approaches to analgesic drug discovery and repurposing and highlight the potential of computational functional genomics in this field including a demonstration of the workflow using a novel R library ‘dbtORA’.
Collapse
Affiliation(s)
- Catharina Lippmann
- Fraunhofer Institute of Molecular Biology & Applied Ecology – Project Group Translational Medicine & Pharmacology (IME–TMP), Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Dario Kringel
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Alfred Ultsch
- DataBionics Research Group, University of Marburg, Hans-Meerwein-Straße 6, 35032 Marburg, Germany
| | - Jörn Lötsch
- Fraunhofer Institute of Molecular Biology & Applied Ecology – Project Group Translational Medicine & Pharmacology (IME–TMP), Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
11
|
Maldonado R, Baños JE, Cabañero D. Usefulness of knockout mice to clarify the role of the opioid system in chronic pain. Br J Pharmacol 2018; 175:2791-2808. [PMID: 29124744 DOI: 10.1111/bph.14088] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022] Open
Abstract
Several lines of knockout mice deficient in the genes encoding each component of the endogenous opioid system have been used for decades to clarify the specific role of the different opioid receptors and peptide precursors in many physiopathological conditions. The use of these genetically modified mice has improved our knowledge of the specific involvement of each endogenous opioid component in nociceptive transmission during acute and chronic pain conditions. The present review summarizes the recent advances obtained using these genetic tools in understanding the role of the opioid system in the pathophysiological mechanisms underlying chronic pain. Behavioural data obtained in these chronic pain models are discussed considering the peculiarities of the behavioural phenotype of each line of knockout mice. These studies have identified the crucial role of specific components of the opioid system in different manifestations of chronic pain and have also opened new possible therapeutic approaches, such as the development of opioid compounds simultaneously targeting several opioid receptors. However, several questions still remain open and require further experimental effort to be clarified. The novel genetic tools now available to manipulate specific neuronal populations and precise genome editing in mice will facilitate in a near future the elucidation of the role of each component of the endogenous opioid system in chronic pain. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
- Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Josep Eladi Baños
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
12
|
Ferdousi M, Finn DP. Stress-induced modulation of pain: Role of the endogenous opioid system. PROGRESS IN BRAIN RESEARCH 2018; 239:121-177. [DOI: 10.1016/bs.pbr.2018.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
13
|
Adem A, Madjid N, Kahl U, Holst S, Sadek B, Sandin J, Terenius L, Ögren SO. Nociceptin and the NOP receptor in aversive learning in mice. Eur Neuropsychopharmacol 2017; 27:1298-1307. [PMID: 29102248 DOI: 10.1016/j.euroneuro.2017.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 09/13/2017] [Accepted: 09/25/2017] [Indexed: 11/27/2022]
Abstract
The endogenous neuropeptide nociceptin (N/OFQ), which mediates its actions via the nociceptin receptor (NOP), is implicated in multiple behavioural and physiological functions. This study examined the effects of the NOP agonists N/OFQ and the synthetic agonist Ro 64-6198, the antagonists NNN and NalBzoH, as well as deletion of the Pronociceptin gene on emotional memory in mice. The animals were tested in the passive avoidance (PA) task, dependent on hippocampal and amygdala functions. N/OFQ injected intraventricularly (i.c.v.) prior to training produced a biphasic effect on PA retention; facilitation at a low dose and impairment at higher doses. Ro 64-6198 also displayed a biphasic effect with memory facilitation at lower doses and impairment at a high dose. None of the agonists influenced PA training latencies. NNN did not significantly modulate retention in the PA task but antagonized the inhibitory effects of N/OFQ. NalBzoH facilitated memory retention in a dose-dependent manner and blocked the impairing effects of N/OFQ. However, neither NNN nor NalBzoH blocked the memory-impairing effects of Ro 64-6198. Finally, the Pnoc knockout mice exhibited enhanced PA retention latencies compared to the wild type mice. The biphasic effect of the natural ligand and Ro 64-6198 and the failure of the antagonists to block the action of Ro 64-6198 indicate complexity in ligand-receptor interaction. These results indicate that brain nociceptin and its NOP has a subtle role in regulation of mechanisms of relevance for treatment of disorders with processing disturbances of aversive events e.g. Alzheimer's disease, anxiety, depression and PTSD.
Collapse
Affiliation(s)
- Abdu Adem
- Department of Neuroscience, Retzius väg 8, S-171 77 Stockholm, Sweden.
| | - Nather Madjid
- Department of Pharmacology & Therapeutics Faculty of Medicine & Health Sciences UAE University, Al Ain, UAE; Department of Neuroscience, Retzius väg 8, S-171 77 Stockholm, Sweden
| | - Ulrika Kahl
- Department of Pharmacology & Therapeutics Faculty of Medicine & Health Sciences UAE University, Al Ain, UAE
| | - Sarah Holst
- Department of Pharmacology & Therapeutics Faculty of Medicine & Health Sciences UAE University, Al Ain, UAE
| | - Bassem Sadek
- Department of Neuroscience, Retzius väg 8, S-171 77 Stockholm, Sweden
| | - Johan Sandin
- AlzeCure Foundation, Karolinska Institutet Science Park, Hälsovägen 7, S-141 57 Huddinge, Stockholm, Sweden
| | - Lars Terenius
- Department of Clinical Neuroscience, Karolinska Institutet, CMM L8:01, Karolinska University Hospital, S-171 76 Stockholm, Sweden
| | - Sven Ove Ögren
- Department of Pharmacology & Therapeutics Faculty of Medicine & Health Sciences UAE University, Al Ain, UAE.
| |
Collapse
|
14
|
Stockdale DP, Titunick MB, Biegler JM, Reed JL, Hartung AM, Wiemer DF, McLaughlin PJ, Neighbors JD. Selective opioid growth factor receptor antagonists based on a stilbene isostere. Bioorg Med Chem 2017; 25:4464-4474. [PMID: 28693915 PMCID: PMC5567982 DOI: 10.1016/j.bmc.2017.06.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/13/2017] [Accepted: 06/21/2017] [Indexed: 02/01/2023]
Abstract
As part of an ongoing drug development effort aimed at selective opioid receptor ligands based on the pawhuskin natural products we have synthesized a small set of amide isosteres. These amides were centered on lead compounds which are selective antagonists for the delta and kappa opioid receptors. The amide isomers revealed here show dramatically different activity from the parent stilbene compounds. Three of the isomers synthesized showed antagonist activity for the opioid growth factor (OGF)/opioid growth factor receptor (OGFR) axis which is involved in cellular and organ growth control. This cellular signaling mechanism is targeted by "low-dose" naltrexone therapy which is being tested clinically for multiple sclerosis, Crohn's disease, cancer, and wound healing disorders. The compounds described here are the first selective small molecule ligands for the OGF/OGFR system and will serve as important leads and probes for further study.
Collapse
Affiliation(s)
- David P Stockdale
- Department of Chemistry, The University of Iowa, Iowa City, IA 52242-1294, United States
| | - Michelle B Titunick
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Jessica M Biegler
- Department of Pharmacology and Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Pennsylvania State University Cancer Institute, Hershey, PA 17033, United States
| | - Jessie L Reed
- Department of Pharmacology and Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Pennsylvania State University Cancer Institute, Hershey, PA 17033, United States
| | - Alyssa M Hartung
- Department of Chemistry, The University of Iowa, Iowa City, IA 52242-1294, United States
| | - David F Wiemer
- Department of Chemistry, The University of Iowa, Iowa City, IA 52242-1294, United States
| | - Patricia J McLaughlin
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Pennsylvania State University Cancer Institute, Hershey, PA 17033, United States
| | - Jeffrey D Neighbors
- Department of Pharmacology and Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Pennsylvania State University Cancer Institute, Hershey, PA 17033, United States.
| |
Collapse
|
15
|
Spaziano G, Sorrentino R, Matteis M, Malgieri G, Sgambato M, Russo TP, Terlizzi M, Roviezzo F, Rossi F, Pinto A, Fattorusso R, D'Agostino B. Nociceptin reduces the inflammatory immune microenvironment in a conventional murine model of airway hyperresponsiveness. Clin Exp Allergy 2017; 47:208-216. [PMID: 27562660 DOI: 10.1111/cea.12808] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/25/2016] [Accepted: 08/19/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Nociceptin/orphanin FQ (N/OFQ) and its receptor (NOP) are involved in airway hyperresponsiveness (AHR) and inflammation. However, the role of nociceptin at modulating the inflammatory immune microenvironment in asthma is still unclear. OBJECTIVE To understand the role of N/OFQ in the regulation of a Th2-like environment, we used a conventional murine model of AHR. METHODS Balb/c and CD1 mice were sensitized to ovalbumin (OVA) and treated with saline solution or N/OFQ, at days 0 and 7. A group of Balb/c mice were killed at 7 and 14 days from the first sensitization for the inflammatory profile evaluation while a group of Balb/c and CD1 mice were aerosol-challenged from day 21 to 23 with OVA and killed 24 h later for functional evaluations. RESULTS In OVA-sensitized mice, N/OFQ significantly reduced IL-4+ CD4+ T cells in lymph nodes (LN) and IL-13 in the lungs, while it induced IFN-γ increase in the lung. The efflux of dendritic cells (DCs) to the mediastinic LN and into the lung of OVA-sensitized mice was reduced in N/OFQ-treated and sensitized mice. N/OFQ reduced the expression of CD80 on DCs, indicating its ability to modulate the activation of DCs. In a less prone Th2-like environment mice strain, such as CD1 mice, N/OFQ did not modify lung resistances as observed in BALB/c mice. Finally, spectroscopic data showed the N/OFQ was able to interact onto the membrane of DCs obtained from Balb/c rather than CD1 mice, indicating its ability to modulate AHR in a Th2-like environment with a direct activity on DCs. CONCLUSIONS AND CLINICAL RELEVANCE Our data confirmed the capability of N/OFQ to modulate the immune microenvironment in the lung of Th2-biased, OVA-sensitized Balb/c mice, suggesting N/OFQ-NOP axis as a novel pharmacological tool to modulate the inflammatory immune microenvironment in asthma.
Collapse
Affiliation(s)
- G Spaziano
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - R Sorrentino
- Department of Pharmacy (DIFARMA), University of Salerno, Fisciano, Italy
| | - M Matteis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - G Malgieri
- Department of Environmental, Biological and Pharmaceutical Science and Technology, Second University of Naples, Caserta, Italy
| | - M Sgambato
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - T P Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - M Terlizzi
- Department of Pharmacy (DIFARMA), University of Salerno, Fisciano, Italy
| | - F Roviezzo
- Department of Experimental Pharmacology, University Federico II of Naples, Naples, Italy
| | - F Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - A Pinto
- Department of Pharmacy (DIFARMA), University of Salerno, Fisciano, Italy
| | - R Fattorusso
- Department of Environmental, Biological and Pharmaceutical Science and Technology, Second University of Naples, Caserta, Italy
| | - B D'Agostino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| |
Collapse
|
16
|
Van Camp KA, Baggerman G, Blust R, Husson SJ. Peptidomics of the zebrafish Danio rerio : In search for neuropeptides. J Proteomics 2017; 150:290-296. [DOI: 10.1016/j.jprot.2016.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/07/2016] [Accepted: 09/27/2016] [Indexed: 12/27/2022]
|
17
|
Anand P, Yiangou Y, Anand U, Mukerji G, Sinisi M, Fox M, McQuillan A, Quick T, Korchev YE, Hein P. Nociceptin/orphanin FQ receptor expression in clinical pain disorders and functional effects in cultured neurons. Pain 2016; 157:1960-1969. [PMID: 27127846 DOI: 10.1097/j.pain.0000000000000597] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The nociceptin/orphanin FQ peptide receptor (NOP), activated by its endogenous peptide ligand nociceptin/orphanin FQ (N/OFQ), exerts several effects including modulation of pain signalling. We have examined, for the first time, the tissue distribution of the NOP receptor in clinical visceral and somatic pain disorders by immunohistochemistry and assessed functional effects of NOP and μ-opioid receptor activation in cultured human and rat dorsal root ganglion (DRG) neurons. Quantification of NOP-positive nerve fibres within the bladder suburothelium revealed a remarkable several-fold increase in detrusor overactivity (P < 0.0001) and painful bladder syndrome patient specimens (P = 0.0014) compared with controls. In postmortem control human DRG, 75% to 80% of small/medium neurons (≤50 μm diameter) in the lumbar (somatic) and sacral (visceral) DRG were positive for NOP, and fewer large neurons; avulsion-injured cervical human DRG neurons showed similar numbers. NOP immunoreactivity was significantly decreased in injured peripheral nerves (P = 0.0004), and also in painful neuromas (P = 0.025). Calcium-imaging studies in cultured rat DRG neurons demonstrated dose-dependent inhibition of capsaicin responses in the presence of N/OFQ, with an IC50 of 8.6 pM. In cultured human DRG neurons, 32% inhibition of capsaicin responses was observed in the presence of 1 pM N/OFQ (P < 0.001). The maximum inhibition of capsaicin responses was greater with N/OFQ than μ-opioid receptor agonist DAMGO. Our findings highlight the potential of NOP agonists, particularly in urinary bladder overactivity and pain syndromes. The regulation of NOP expression in visceral and somatic sensory neurons by target-derived neurotrophic factors deserves further study, and the efficacy of NOP selective agonists in clinical trials.
Collapse
Affiliation(s)
- Praveen Anand
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Yiangos Yiangou
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Uma Anand
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Gaurav Mukerji
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Marco Sinisi
- Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Michael Fox
- Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Anthony McQuillan
- Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Tom Quick
- Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Yuri E Korchev
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Peter Hein
- Grünenthal Innovation, Translational Science and Strategy, Grünenthal GmbH, Aachen, Germany
| |
Collapse
|
18
|
Matsushima A, Nishimura H, Matsuyama Y, Liu X, Costa T, Shimohigashi Y. Specific affinity-labeling of the nociceptin ORL1 receptor using a thiol-activated Cys(Npys)-containing peptide ligand. Biopolymers 2016; 106:460-9. [DOI: 10.1002/bip.22792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/22/2015] [Accepted: 11/02/2015] [Indexed: 11/05/2022]
Affiliation(s)
- Ayami Matsushima
- Department of Chemistry, Laboratory of Structure-Function Biochemistry, Faculty and Graduate School of Science; Kyushu University; Fukuoka 819-0395 Japan
| | - Hirokazu Nishimura
- Department of Chemistry, Laboratory of Structure-Function Biochemistry, Faculty and Graduate School of Science; Kyushu University; Fukuoka 819-0395 Japan
| | - Yutaka Matsuyama
- Department of Chemistry, Laboratory of Structure-Function Biochemistry, Faculty and Graduate School of Science; Kyushu University; Fukuoka 819-0395 Japan
| | - Xiaohui Liu
- Department of Chemistry, Laboratory of Structure-Function Biochemistry, Faculty and Graduate School of Science; Kyushu University; Fukuoka 819-0395 Japan
| | - Tommaso Costa
- Istituto Superiore Di Sanità; Laboratorio Di Farmacologia; Viale Regina Elena 299 Roma Italy
| | - Yasuyuki Shimohigashi
- Department of Chemistry, Laboratory of Structure-Function Biochemistry, Faculty and Graduate School of Science; Kyushu University; Fukuoka 819-0395 Japan
| |
Collapse
|
19
|
The nociceptin/orphanin FQ receptor agonist SR-8993 as a candidate therapeutic for alcohol use disorders: validation in rat models. Psychopharmacology (Berl) 2016; 233:3553-63. [PMID: 27515665 PMCID: PMC5021736 DOI: 10.1007/s00213-016-4385-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/16/2016] [Indexed: 12/30/2022]
Abstract
RATIONALE Alcoholism is a complex disorder in which diverse pathophysiological processes contribute to initiation and progression, resulting in a high degree of heterogeneity among patients. Few pharmacotherapies are presently available, and patient responses to these are variable. The nociceptin/orphanin FQ (NOP) receptor has been suggested to play a role both in alcohol reward and in negatively reinforced alcohol seeking. Previous studies have shown that NOP-receptor activation reduces alcohol intake in genetically selected alcohol-preferring as well as alcohol-dependent rats. NOP activation also blocks stress- and cue-induced reinstatement of alcohol-seeking behavior. OBJECTIVES Here, we aimed to examine a novel, potent, and brain-penetrant small-molecule NOP-receptor agonist, SR-8993, in animal models of alcohol- as well as anxiety-related behavior using male Wistar rats. RESULTS SR-8993 was mildly anxiolytic when given to naïve animals and potently reversed acute alcohol withdrawal-induced ("hangover") anxiety. SR-8993 reduced both home-cage limited access drinking, operant responding for alcohol, and escalation induced through prolonged intermittent access to alcohol. SR-8993 further attenuated stress- as well as cue-induced relapse to alcohol seeking. For the effective dose (1.0 mg/kg), non-specific effects such as sedation may be limited, since a range of control behaviors were unaffected, and this dose did not interact with alcohol elimination. CONCLUSION These findings provide further support for NOP-receptor agonism as a promising candidate treatment for alcoholism and establish SR-8993 or related molecules as suitable for further development as therapeutics.
Collapse
|
20
|
Andero R. Nociceptin and the nociceptin receptor in learning and memory. Prog Neuropsychopharmacol Biol Psychiatry 2015; 62:45-50. [PMID: 25724763 PMCID: PMC4458422 DOI: 10.1016/j.pnpbp.2015.02.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 11/21/2022]
Abstract
There are many processes in which the neuropeptide nociceptin/orphanin FQ (N/OFQ or nociceptin) is involved in the brain. The role of nociceptin in learning and memory holds promise in modulating these processes in health and disease in the human brain. This review summarizes the body of research focused on N/OFQ and its specific receptor, the nociceptin receptor (NOP receptor), in learning and memory, and its potential mechanisms of action, in which acetylcholine, NMDA receptor, and noradrenaline may be critical. Finally, the association between NOP receptor and posttraumatic stress disorder (PTSD), a psychiatric disorder with altered fear learning, is examined as one of the potential outcomes resulting from pathological consequences of dysregulation of N/OFQ-NOP receptor in the brain.
Collapse
Affiliation(s)
- Raül Andero
- Department of Psychiatry and Behavioral Sciences, Center for Behavioral Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA.
| |
Collapse
|
21
|
Wang PC, Ho IK, Lee CWS. Buprenorphine-elicited alteration of adenylate cyclase activity in human embryonic kidney 293 cells coexpressing κ-, μ-opioid and nociceptin receptors. J Cell Mol Med 2015; 19:2587-96. [PMID: 26153065 PMCID: PMC4627564 DOI: 10.1111/jcmm.12644] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/28/2015] [Indexed: 02/03/2023] Open
Abstract
Buprenorphine, a maintenance drug for heroin addicts, exerts its pharmacological function via κ- (KOP), μ-opioid (MOP) and nociceptin/opioid receptor-like 1 (NOP) receptors. Previously, we investigated its effects in an in vitro model expressing human MOP and NOP receptors individually or simultaneously (MOP, NOP, and MOP+NOP) in human embryonic kidney 293 cells. Here, we expanded this cell model by expressing human KOP, MOP and NOP receptors individually or simultaneously (KOP, KOP+MOP, KOP+NOP and KOP+MOP+NOP). Radioligand binding with tritium-labelled diprenorphine confirmed the expression of KOP receptors. Immunoblotting and immunocytochemistry indicated that the expressed KOP, MOP and NOP receptors are N-linked glycoproteins and colocalized in cytoplasmic compartments. Acute application of the opioid receptor agonists— U-69593, DAMGO and nociceptin— inhibited adenylate cyclase (AC) activity in cells expressing KOP, MOP and NOP receptors respectively. Buprenorphine, when applied acutely, inhibited AC activity to ~90% in cells expressing KOP+MOP+NOP receptors. Chronic exposure to buprenorphine induced concentration-dependent AC superactivation in cells expressing KOP+NOP receptors, and the level of this superactivation was even higher in KOP+MOP+NOP-expressing cells. Our study demonstrated that MOP receptor could enhance AC regulation in the presence of coexpressed KOP and NOP receptors, and NOP receptor is essential for concentration-dependent AC superactivation elicited by chronic buprenorphine exposure.
Collapse
Affiliation(s)
- Pei-Chen Wang
- Neuropsychiatric Center, National Health Research Institutes, Miaoli County, Taiwan
| | - Ing-Kang Ho
- Neuropsychiatric Center, National Health Research Institutes, Miaoli County, Taiwan.,Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Center for Drug Abuse and Addiction, China Medical University Hospital, Taichung, Taiwan
| | - Cynthia Wei-Sheng Lee
- Center for Drug Abuse and Addiction, China Medical University Hospital, Taichung, Taiwan.,China Medical University, Taichung, Taiwan
| |
Collapse
|
22
|
Sacco E, Recupero S, Bientinesi R, Palermo G, D’Agostino D, Currò D, Bassi P. Pioneering drugs for overactive bladder and detrusor overactivity: Ongoing research and future directions. World J Obstet Gynecol 2015; 4:24-39. [DOI: 10.5317/wjog.v4.i2.24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 01/31/2015] [Accepted: 04/14/2015] [Indexed: 02/05/2023] Open
Abstract
The ongoing research on pioneering drug candidates for the overactive bladder (OAB) aimed to overcome the limitations of currently licensed pharmacotherapies, such as antimuscarinics, β3-adrenergic agents, and botulinum neurotoxin, has been reviewed performing a systematic literature review and web search. The review covers the exploratory agents alternative to available medications for OAB and that may ultimately prove to be therapeutically useful in the future management of OAB patients based on preclinical and early clinical data. It emerges that many alternative pharmacological strategies have been discovered or are under investigation in disease-oriented studies. Several potential therapeutics are known for years but still find obstacles to pass the clinical stages of development, while other completely novel compounds, targeting new pharmacological targets, have been recently discovered and show potential to translate into clinical therapeutic agents for idiopathic and neurogenic OAB syndrome. The global scenario of investigational drugs for OAB gives promise for the development of innovative therapeutics that may ultimately prove effective as first, combined or second-line treatments within a realistic timescale of ten years.
Collapse
|
23
|
Hallberg M. Neuropeptides: metabolism to bioactive fragments and the pharmacology of their receptors. Med Res Rev 2015; 35:464-519. [PMID: 24894913 DOI: 10.1002/med.21323] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
The proteolytic processing of neuropeptides has an important regulatory function and the peptide fragments resulting from the enzymatic degradation often exert essential physiological roles. The proteolytic processing generates, not only biologically inactive fragments, but also bioactive fragments that modulate or even counteract the response of their parent peptides. Frequently, these peptide fragments interact with receptors that are not recognized by the parent peptides. This review discusses tachykinins, opioid peptides, angiotensins, bradykinins, and neuropeptide Y that are present in the central nervous system and their processing to bioactive degradation products. These well-known neuropeptide systems have been selected since they provide illustrative examples that proteolytic degradation of parent peptides can lead to bioactive metabolites with different biological activities as compared to their parent peptides. For example, substance P, dynorphin A, angiotensin I and II, bradykinin, and neuropeptide Y are all degraded to bioactive fragments with pharmacological profiles that differ considerably from those of the parent peptides. The review discusses a selection of the large number of drug-like molecules that act as agonists or antagonists at receptors of neuropeptides. It focuses in particular on the efforts to identify selective drug-like agonists and antagonists mimicking the effects of the endogenous peptide fragments formed. As exemplified in this review, many common neuropeptides are degraded to a variety of smaller fragments but many of the fragments generated have not yet been examined in detail with regard to their potential biological activities. Since these bioactive fragments contain a small number of amino acid residues, they provide an ideal starting point for the development of drug-like substances with ability to mimic the effects of the degradation products. Thus, these substances could provide a rich source of new pharmaceuticals. However, as discussed herein relatively few examples have so far been disclosed of successful attempts to create bioavailable, drug-like agonists or antagonists, starting from the structure of endogenous peptide fragments and applying procedures relying on stepwise manipulations and simplifications of the peptide structures.
Collapse
Affiliation(s)
- Mathias Hallberg
- Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, Biomedical Center, Uppsala, Sweden
| |
Collapse
|
24
|
Mechanical hyperalgesia in rats with diabetic polyneuropathy is selectively inhibited by local peripheral nociceptin/orphanin FQ receptor and µ-opioid receptor agonism. Eur J Pharmacol 2015; 754:61-5. [DOI: 10.1016/j.ejphar.2015.01.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 01/08/2015] [Accepted: 01/26/2015] [Indexed: 01/31/2023]
|
25
|
Baiula M, Bedini A, Spampinato SM. Role of nociceptin/orphanin FQ in thermoregulation. Neuropeptides 2015; 50:51-6. [PMID: 25812480 DOI: 10.1016/j.npep.2015.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 02/25/2015] [Accepted: 03/11/2015] [Indexed: 01/30/2023]
Abstract
Nociceptin/Orphanin FQ (N/OFQ) is a 17-amino acid peptide that binds to the nociceptin receptor (NOP). N/OFQ and NOP receptors are expressed in numerous brain areas. The generation of specific agonists, antagonists and receptor-deficient mice or rats has enabled progress in elucidating the biological functions of N/OFQ. These tools have been employed to identify the biological significance of the N/OFQ system and how it interacts with other endogenous systems to regulate several body functions. The present review focuses on the role of N/OFQ in the regulation of body temperature and its relationship with energy balance. Critical evaluation of the literature data suggests that N/OFQ, acting through the NOP receptor, may cause hypothermia by influencing the complex thermoregulatory system that operates as a federation of independent thermoeffector loops to control body temperature at the hypothalamic level. Furthermore, N/OFQ counteracts hyperthermia elicited by cannabinoids or µ-opioid agonists. N/OFQ-induced hypothermia is prevented by ω-conotoxin GVIA, an N-type calcium channel blocker. Hypothermia induced by N/OFQ is considered within the framework of the complex action that this neuropeptide exerts on energy balance. Energy stores are regulated through the complex neural controls exerted on both food intake and energy expenditure. In laboratory rodents, N/OFQ stimulates consummatory behavior and decreases energy expenditure. Taken together, these studies support the idea that N/OFQ contributes to the regulation of energy balance by acting as an "anabolic" neuropeptide as it elicits effects similar to those produced in the hypothalamus by other neuropeptides such as orexins and neuropeptide Y.
Collapse
Affiliation(s)
- Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, Italy
| | - Andrea Bedini
- Department of Pharmacy and Biotechnology, University of Bologna, Italy
| | | |
Collapse
|
26
|
Stevens CW. Bioinformatics and evolution of vertebrate nociceptin and opioid receptors. VITAMINS AND HORMONES 2015; 97:57-94. [PMID: 25677768 DOI: 10.1016/bs.vh.2014.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
G protein-coupled receptors (GPCRs) are ancestrally related membrane proteins on cells that mediate the pharmacological effect of most drugs and neurotransmitters. GPCRs are the largest group of membrane receptor proteins encoded in the human genome. One of the most famous types of GPCRs is the opioid receptors. Opioid family receptors consist of four closely related proteins expressed in all vertebrate brains and spinal cords examined to date. The three classical types of opioid receptors shown unequivocally to mediate analgesia in animal models and in humans are the mu- (MOR), delta- (DOR), and kappa-(KOR) opioid receptor proteins. The fourth and most recent member of the opioid receptor family discovered is the nociceptin or orphanin FQ receptor (ORL). The role of ORL and its ligands in producing analgesia is not as clear, with both analgesic and hyperalgesic effects reported. All four opioid family receptor genes were cloned from expressed mRNA in a number of vertebrate species, and there are enough sequences presently available to carry out bioinformatic analysis. This chapter presents the results of a comparative analysis of vertebrate opioid receptors using pharmacological studies, bioinformatics, and the latest data from human whole-genome studies. Results confirm our initial hypotheses that the four opioid receptor genes most likely arose by whole-genome duplication, that there is an evolutionary vector of opioid receptor type divergence in sequence and function, and that the hMOR gene shows evidence of positive selection or adaptive evolution in Homo sapiens.
Collapse
Affiliation(s)
- Craig W Stevens
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma, USA.
| |
Collapse
|
27
|
Corrado S, Battisti UM, Sorbi C, Tait A, Malfacini D, Camarda V, Calò G, Brasili L. Synthesis and Structure-Activity Relationships of Triazaspirodecanone Derivatives as Nociceptin/Orphanin FQ Receptor Ligands. Chem Biol Drug Des 2015; 86:447-58. [DOI: 10.1111/cbdd.12505] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/17/2014] [Accepted: 12/15/2014] [Indexed: 11/27/2022]
Affiliation(s)
- Sandra Corrado
- Department of Life Sciences; University of Modena & Reggio Emilia; Via G. Campi 183 41125 Modena Italy
| | - Umberto M. Battisti
- Department of Life Sciences; University of Modena & Reggio Emilia; Via G. Campi 183 41125 Modena Italy
| | - Claudia Sorbi
- Department of Life Sciences; University of Modena & Reggio Emilia; Via G. Campi 183 41125 Modena Italy
| | - Annalisa Tait
- Department of Life Sciences; University of Modena & Reggio Emilia; Via G. Campi 183 41125 Modena Italy
| | - Davide Malfacini
- Department of Medical Sciences; Section of Pharmacology and National Institute of Neuroscience; University of Ferrara; 44121 Ferrara Italy
| | - Valeria Camarda
- Department of Medical Sciences; Section of Pharmacology and National Institute of Neuroscience; University of Ferrara; 44121 Ferrara Italy
| | - Girolamo Calò
- Department of Medical Sciences; Section of Pharmacology and National Institute of Neuroscience; University of Ferrara; 44121 Ferrara Italy
| | - Livio Brasili
- Department of Life Sciences; University of Modena & Reggio Emilia; Via G. Campi 183 41125 Modena Italy
| |
Collapse
|
28
|
Xie XS. The neuronal circuit between nociceptin/orphanin FQ and hypocretins/orexins coordinately modulates stress-induced analgesia and anxiety-related behavior. VITAMINS AND HORMONES 2015; 97:295-321. [PMID: 25677777 DOI: 10.1016/bs.vh.2014.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
The neuropeptide nociceptin/orphanin FQ (N/OFQ), acting on its receptors (NOP), modulates a variety of biological functions and neurobehavior including nociception, stress responses, water and food-intake, locomotor activity, and spatial attention. N/OFQ is conventionally regarded as an "antiopiate" peptide in the brain because central administration of N/OFQ attenuates stress-induced analgesia (SIA) and produces anxiolytic effects. However, naloxone-irreversible SIA and anxiolytic action are unlikely to be mediated by the opiate system. Both N/OFQ and NOP receptors are expressed most abundantly in the hypothalamus, where two other neuropeptides, the hypocretins/orexins (Hcrts), are exclusively synthesized in the lateral hypothalamic area. N/OFQ and Hcrt regulate most cellular physiological responses in opposite directions (e.g., ion channel modulation and second messenger coupling), and produce differential modulations for almost all neurobehavior assessed, including sleep/wake, locomotion, and rewarding behaviors. This chapter focuses on recent studies that provide evidence at a neuroanatomical level showing that a local neuronal circuit linking N/OFQ to Hcrt neurons exists. Functionally, N/OFQ depresses Hcrt neuronal activity at the cellular level, and modulates stress responses, especially SIA and anxiety-related behavior in the whole organism. N/OFQ exerts its attenuation of SIA and anxiolytic action on fear-induced anxiety through direct modulation of Hcrt neuronal activity. The information obtained from these studies has provided insights into how interaction between the Hcrt and N/OFQ systems positively and negatively modulates the complex and integrated stress responses.
Collapse
Affiliation(s)
- Xinmin Simon Xie
- AfaSci Research Laboratories, Redwood City, California, USA; Department of Anesthesia, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
29
|
Gavioli EC, de Medeiros IU, Monteiro MC, Calo G, Romão PRT. Nociceptin/orphanin FQ-NOP receptor system in inflammatory and immune-mediated diseases. VITAMINS AND HORMONES 2015; 97:241-66. [PMID: 25677775 DOI: 10.1016/bs.vh.2014.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neuropeptide nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand of the G-protein-coupled receptor NOP. Cells from the immune system express the precursor preproN/OFQ and the NOP receptor, as well as secrete N/OFQ. The activation of the N/OFQ-NOP pathway can regulate inflammatory and immune responses. Several immune activities, including leukocyte migration, cytokine and chemokine production, and lymphocytes proliferation are influenced by NOP activation. It was demonstrated that cytokines and other stimuli such as Toll-like receptor agonist (e.g., lipopolysaccharide) induce N/OFQ production by cells from innate and adaptive immune response. In this context, N/OFQ could modulate the outcome of inflammatory diseases, such as sepsis and immune-mediated pathologies by mechanisms not clearly elucidated. In fact, clinical studies revealed increased levels of N/OFQ under sepsis, arthritis, and Parkinson's disease. Preclinical and clinical studies pointed to the blockade of NOP receptor signaling as successful strategy for the treatment of inflammatory diseases. This review is focused on experimental and clinical data that suggest the participation of N/OFQ-NOP receptor activation in the modulation of the immune response, highlighting the immunomodulatory potential of NOP antagonists in the inflammatory and immunological disturbances.
Collapse
Affiliation(s)
- Elaine C Gavioli
- Department of Biophysic and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Iris Ucella de Medeiros
- Department of Biophysic and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Marta C Monteiro
- Laboratory of Clinical Microbiology and Immunology, Faculty of Pharmacy, Federal University of Pará, Belém, Brazil
| | - Girolamo Calo
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Pedro R T Romão
- Laboratory of Immunology, Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre, Rua Sarmento Leite, Porto Alegre, Brazil.
| |
Collapse
|
30
|
Lohman RJ, Harrison RS, Ruiz-Gómez G, Hoang HN, Shepherd NE, Chow S, Hill TA, Madala PK, Fairlie DP. Helix-constrained nociceptin peptides are potent agonists and antagonists of ORL-1 and nociception. VITAMINS AND HORMONES 2015; 97:1-55. [PMID: 25677767 DOI: 10.1016/bs.vh.2014.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nociceptin (orphanin FQ) is a 17-residue neuropeptide hormone with roles in both nociception and analgesia. It is an opioid-like peptide that binds to and activates the G-protein-coupled receptor opioid receptor-like-1 (ORL-1, NOP, orphanin FQ receptor, kappa-type 3 opioid receptor) on central and peripheral nervous tissue, without activating classic delta-, kappa-, or mu-opioid receptors or being inhibited by the classic opioid antagonist naloxone. The three-dimensional structure of ORL-1 was recently published, and the activation mechanism is believed to involve capture by ORL-1 of the high-affinity binding, prohelical C-terminus. This likely anchors the receptor-activating N-terminus of nociception nearby for insertion in the membrane-spanning helices of ORL-1. In search of higher agonist potency, two lysine and two aspartate residues were strategically incorporated into the receptor-binding C-terminus of the nociceptin sequence and two Lys(i)→Asp(i+4) side chain-side chain condensations were used to generate lactam cross-links that constrained nociceptin into a highly stable α-helix in water. A cell-based assay was developed using natively expressed ORL-1 receptors on mouse neuroblastoma cells to measure phosphorylated ERK as a reporter of agonist-induced receptor activation and intracellular signaling. Agonist activity was increased up to 20-fold over native nociceptin using a combination of this helix-inducing strategy and other amino acid modifications. An NMR-derived three-dimensional solution structure is described for a potent ORL-1 agonist derived from nociceptin, along with structure-activity relationships leading to the most potent known α-helical ORL-1 agonist (EC₅₀ 40 pM, pERK, Neuro-2a cells) and antagonist (IC₅₀ 7 nM, pERK, Neuro-2a cells). These α-helix-constrained mimetics of nociceptin(1-17) had enhanced serum stability relative to unconstrained peptide analogues and nociceptin itself, were not cytotoxic, and displayed potent thermal analgesic and antianalgesic properties in rats (ED₅₀ 70 pmol, IC₅₀ 10 nmol, s.c.), suggesting promising uses in vivo for the treatment of pain and other ORL-1-mediated responses.
Collapse
Affiliation(s)
- Rink-Jan Lohman
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Rosemary S Harrison
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Gloria Ruiz-Gómez
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Huy N Hoang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas E Shepherd
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Shiao Chow
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Timothy A Hill
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Praveen K Madala
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
31
|
Rutten K, Tzschentke TM, Koch T, Schiene K, Christoph T. Pharmacogenomic study of the role of the nociceptin/orphanin FQ receptor and opioid receptors in diabetic hyperalgesia. Eur J Pharmacol 2014; 741:264-71. [PMID: 25169429 DOI: 10.1016/j.ejphar.2014.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 07/10/2014] [Accepted: 08/05/2014] [Indexed: 01/03/2023]
Abstract
Targeting functionally independent receptors may provide synergistic analgesic effects in neuropathic pain. To examine the interdependency between different opioid receptors (µ-opioid peptide [MOP], δ-opioid peptide [DOP] and κ-opioid peptide [KOP]) and the nociceptin/orphanin FQ peptide (NOP) receptor in streptozotocin (STZ)-induced diabetic polyneuropathy, nocifensive activity was measured using a hot plate test in wild-type and NOP, MOP, DOP and KOP receptor knockout mice in response to the selective receptor agonists Ro65-6570, morphine, SNC-80 and U50488H, or vehicle. Nocifensive activity was similar in non-diabetic wild-type and knockout mice at baseline, before agonist or vehicle administration. STZ-induced diabetes significantly increased heat sensitivity in all mouse strains, but MOP, DOP and KOP receptor knockouts showed a smaller degree of hyperalgesia than wild-type mice and NOP receptor knockouts. For each agonist, a significant antihyperalgesic effect was observed in wild-type diabetic mice (all P<0.05 versus vehicle); the effect was markedly attenuated in diabetic mice lacking the cognate receptor compared with wild-type diabetic mice. Morphine was the only agonist that demonstrated near-full antihyperalgesic efficacy across all non-cognate receptor knockouts. Partial or near-complete reductions in efficacy were observed with Ro65-6570 in DOP and KOP receptor knockouts, with SNC-80 in NOP, MOP and KOP receptor knockouts, and with U50488H in NOP and DOP receptor knockouts. There was no evidence of NOP and MOP receptor interdependency in response to selective agonists for these receptors. These findings suggest that concurrent activation of NOP and MOP receptors, which showed functional independence, may yield an effective and favorable therapeutic analgesic profile.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/therapeutic use
- Animals
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Hyperalgesia/drug therapy
- Hyperalgesia/genetics
- Hyperalgesia/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid/physiology
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/physiology
- Nociceptin Receptor
Collapse
Affiliation(s)
- Kris Rutten
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Pain Pharmacology, Zieglerstrasse 6, 52078 Aachen, Germany.
| | - Thomas M Tzschentke
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Pain Pharmacology, Zieglerstrasse 6, 52078 Aachen, Germany
| | - Thomas Koch
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Molecular Pharmacology, Aachen, Germany
| | - Klaus Schiene
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Pain Pharmacology, Zieglerstrasse 6, 52078 Aachen, Germany
| | - Thomas Christoph
- Grünenthal GmbH, Global Preclinical Drug Development, Department of Pain Pharmacology, Zieglerstrasse 6, 52078 Aachen, Germany
| |
Collapse
|
32
|
Kallupi M, Varodayan FP, Oleata CS, Correia D, Luu G, Roberto M. Nociceptin/orphanin FQ decreases glutamate transmission and blocks ethanol-induced effects in the central amygdala of naive and ethanol-dependent rats. Neuropsychopharmacology 2014; 39:1081-92. [PMID: 24169802 PMCID: PMC3957102 DOI: 10.1038/npp.2013.308] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/08/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022]
Abstract
The central nucleus of the amygdala (CeA) mediates several addiction-related processes and nociceptin/orphanin FQ (nociceptin) regulates ethanol intake and anxiety-like behaviors. Glutamatergic synapses, in the CeA and throughout the brain, are very sensitive to ethanol and contribute to alcohol reinforcement, tolerance, and dependence. Previously, we reported that in the rat CeA, acute and chronic ethanol exposures significantly decrease glutamate transmission by both pre- and postsynaptic actions. In this study, using electrophysiological techniques in an in vitro CeA slice preparation, we investigated the effects of nociceptin on glutamatergic transmission and its interaction with acute ethanol in naive and ethanol-dependent rats. We found that nociceptin (100-1000 nM) diminished basal-evoked compound glutamatergic receptor-mediated excitatory postsynaptic potentials (EPSPs) and spontaneous and miniature EPSCs (s/mEPSCs) by mainly decreasing glutamate release in the CeA of naive rats. Notably, nociceptin blocked the inhibition induced by acute ethanol (44 mM) and ethanol blocked the nociceptin-induced inhibition of evoked EPSPs in CeA neurons of naive rats. In neurons from chronic ethanol-treated (ethanol-dependent) rats, the nociceptin-induced inhibition of evoked EPSP amplitude was not significantly different from that in naive rats. Application of [Nphe1]Nociceptin(1-13)NH2, a nociceptin receptor (NOP) antagonist, revealed tonic inhibitory activity of NOP on evoked CeA glutamatergic transmission only in ethanol-dependent rats. The antagonist also blocked nociceptin-induced decreases in glutamatergic responses, but did not affect ethanol-induced decreases in evoked EPSP amplitude. Taken together, these studies implicate a potential role for the nociceptin system in regulating glutamatergic transmission and a complex interaction with ethanol at CeA glutamatergic synapses.
Collapse
Affiliation(s)
- Marsida Kallupi
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Florence P Varodayan
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher S Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Diego Correia
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Pharmacology, Universidade Federal do Paraná, Jardim das Américas, Curitiba, Paraná, Brazil
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
33
|
Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW. Calcium-permeable ion channels in pain signaling. Physiol Rev 2014; 94:81-140. [PMID: 24382884 DOI: 10.1152/physrev.00023.2013] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The detection and processing of painful stimuli in afferent sensory neurons is critically dependent on a wide range of different types of voltage- and ligand-gated ion channels, including sodium, calcium, and TRP channels, to name a few. The functions of these channels include the detection of mechanical and chemical insults, the generation of action potentials and regulation of neuronal firing patterns, the initiation of neurotransmitter release at dorsal horn synapses, and the ensuing activation of spinal cord neurons that project to pain centers in the brain. Long-term changes in ion channel expression and function are thought to contribute to chronic pain states. Many of the channels involved in the afferent pain pathway are permeable to calcium ions, suggesting a role in cell signaling beyond the mere generation of electrical activity. In this article, we provide a broad overview of different calcium-permeable ion channels in the afferent pain pathway and their role in pain pathophysiology.
Collapse
|
34
|
Kallupi M, Oleata CS, Luu G, Teshima K, Ciccocioppo R, Roberto M. MT-7716, a novel selective nonpeptidergic NOP receptor agonist, effectively blocks ethanol-induced increase in GABAergic transmission in the rat central amygdala. Front Integr Neurosci 2014; 8:18. [PMID: 24600360 PMCID: PMC3927450 DOI: 10.3389/fnint.2014.00018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/01/2014] [Indexed: 11/13/2022] Open
Abstract
The GABAergic system in the central amygdala (CeA) plays a major role in ethanol dependence and the anxiogenic-like response to ethanol withdrawal. A large body of evidence shows that Nociceptin/Orphanin FQ (N/OFQ) regulates ethanol intake and anxiety-like behavior. In the rat, ethanol significantly augments CeA GABA release, whereas N/OFQ diminishes it. Using electrophysiological techniques in an in vitro slice preparation, in this study we investigated the effects of a nonpeptidergic NOP receptor agonist, MT-7716 [(R)-2-3-[1-(Acenaphthen-1-yl)piperidin-4-yl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl-N-methylacetamide hydrochloride hydrate], and its interaction with ethanol on GABAergic transmission in CeA slices of naïve rats. We found that MT-7716 dose-dependently (100-1000 nM) diminished evoked GABAA receptor-mediated inhibitory postsynaptic potentials (IPSPs) and increased paired-pulse facilitation (PPF) ratio of these evoked IPSPs, suggesting a presynaptic site of action of the MT-7716 by decreasing GABA release at CeA synapses. The presynaptic action of MT-7716 was also supported by the significant decrease in the frequency of miniature inhibitory postsynaptic currents (mIPSCs) induced by the nociceptin receptor (NOP) agonist. Interestingly, MT-7716 prevented the ethanol-induced augmentation of evoked IPSPs. A putative selective NOP antagonist, [Nphe1]Nociceptin(1-13)NH2, totally prevented the MT-7716-induced inhibition of IPSP amplitudes indicating that MT-7716 exerts its effect through NOPs. These data provide support for an interaction between the nociceptin and GABAergic systems in the CeA and for the anti-alcohol properties of the NOP activation. The development of a synthetic nonpeptidergic NOP receptor agonist such as MT-7716 may represent a useful therapeutic target for alcoholism.
Collapse
Affiliation(s)
- Marsida Kallupi
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
- Pharmacology Unit, School of Pharmacy, University of CamerinoCamerino, Italy
| | - Christopher S. Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| | - Koji Teshima
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma CorporationYokohama, Japan
| | - Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of CamerinoCamerino, Italy
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| |
Collapse
|
35
|
Ciccocioppo R, de Guglielmo G, Hansson AC, Ubaldi M, Kallupi M, Cruz MT, Oleata CS, Heilig M, Roberto M. Restraint stress alters nociceptin/orphanin FQ and CRF systems in the rat central amygdala: significance for anxiety-like behaviors. J Neurosci 2014; 34:363-72. [PMID: 24403138 PMCID: PMC3870926 DOI: 10.1523/jneurosci.2400-13.2014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 10/02/2013] [Accepted: 11/02/2013] [Indexed: 02/05/2023] Open
Abstract
Corticotropin releasing factor (CRF) is the primary mediator of stress responses, and nociceptin/orphanin FQ (N/OFQ) plays an important role in the modulation of these stress responses. Thus, in this multidisciplinary study, we explored the relationship between the N/OFQ and the CRF systems in response to stress. Using in situ hybridization (ISH), we assessed the effect of body restraint stress on the gene expression of CRF and N/OFQ-related genes in various subdivisions of the amygdala, a critical brain structure involved in the modulation of stress response and anxiety-like behaviors. We found a selective upregulation of the NOP and downregulation of the CRF1 receptor transcripts in the CeA and in the BLA after body restraint. Thus, we performed intracellular electrophysiological recordings of GABAA-mediated IPSPs in the central nucleus of the amygdala (CeA) to explore functional interactions between CRF and N/OFQ systems in this brain region. Acute application of CRF significantly increased IPSPs in the CeA, and this enhancement was blocked by N/OFQ. Importantly, in stress-restraint rats, baseline CeA GABAergic responses were elevated and N/OFQ exerted a larger inhibition of IPSPs compared with unrestraint rats. The NOP antagonist [Nphe1]-nociceptin(1-13)NH2 increased the IPSP amplitudes in restraint rats but not in unrestraint rats, suggesting a functional recruitment of the N/OFQ system after acute stress. Finally, we evaluated the anxiety-like response in rats subjected to restraint stress and nonrestraint rats after N/OFQ microinjection into the CeA. Intra-CeA injections of N/OFQ significantly and selectively reduced anxiety-like behavior in restraint rats in the elevated plus maze. These combined results demonstrate that acute stress increases N/OFQ systems in the CeA and that N/OFQ has antistress properties.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
| | - Giordano de Guglielmo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
| | - Anita C. Hansson
- Institute of Psychopharmacology at the Central Institute of Mental Health, Medical Faculty Mannheim/University of Heidelberg, Heidelberg 68159, Germany
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
| | - Marsida Kallupi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino 62032, Italy
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, and
| | - Maureen T. Cruz
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, and
| | - Christopher S. Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, and
| | - Markus Heilig
- National Institutes of Health/National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland 20892-1108
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, and
| |
Collapse
|
36
|
Lee H. Effects of co-administration of intrathecal nociceptin/orphanin FQ and opioid antagonists on formalin-induced pain in rats. Yonsei Med J 2013; 54:763-71. [PMID: 23549827 PMCID: PMC3635624 DOI: 10.3349/ymj.2013.54.3.763] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 11/10/2012] [Accepted: 12/12/2012] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Nociceptin/orphanin FQ (N/OFQ) as an endogeneous hexadecapeptide is known to exert antinociceptive effects spinally. The aims of this study were to demonstrate the antinociceptive effects of i.t. N/OFQ and to investigate the possible interaction between N/OFQ and endogenous opioid systems using selective opioid receptor antagonists in rat formalin tests. MATERIALS AND METHODS I.t. N/OFQ was injected in different doses (1-10 nmol) via a lumbar catheter prior to a 50 μL injection of 5% formalin into the right hindpaw of rats. Flinching responses were measured from 0-10 min (phase I, an initial acute state) and 11-60 min (phase II, a prolonged tonic state). To observe which opioid receptors are involved in the anti-nociceptive effect of i.t. N/OFQ in the rat-formalin tests, naltrindole (5-20 nmol), β-funaltrexamine (1-10 nmol), and norbinaltorphimine (10 nmol), selective δ-, μ- and κ-opioid receptor antagonists, respectively, were administered intrathecally 5 min after i.t. N/OFQ. RESULTS I.t. N/OFQ attenuated the formalin-induced flinching responses in a dose-dependent manner in both phases I and II. I.t. administration of naltrindole and β-funaltrexamine dose-dependently reversed the N/OFQ-induced attenuation of flinching responses in both phases; however, norbinaltorphimine did not. CONCLUSION I.t. N/OFQ exerted an antinociceptive effect in both phases of the rat-formalin test through the nociceptin opioid peptide receptor. In addition, the results suggested that δ- and μ-opioid receptors, but not κ-opioid receptors, are involved in the antinociceptive effects of N/OFQ in the spinal cord of rats.
Collapse
Affiliation(s)
- Heeseung Lee
- Department of Anesthesiology and Pain Medicine, School of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul 158-710, Korea.
| |
Collapse
|
37
|
Singh SR, Sullo N, D'Agostino B, Brightling CE, Lambert DG. The effects of nociceptin peptide (N/OFQ)-receptor (NOP) system activation in the airways. Peptides 2013; 39:36-46. [PMID: 23123316 DOI: 10.1016/j.peptides.2012.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 10/17/2012] [Accepted: 10/22/2012] [Indexed: 11/20/2022]
Abstract
The heptadecapeptide nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand for the N/OFQ peptide (NOP) receptor. It is cleaved from a larger precursor identified as prepronociceptin (ppN/OFQ). NOP is a member of the seven transmembrane-spanning G-protein coupled receptor (GPCR) family. ppN/OFQ and NOP receptors are widely distributed in different human tissues. Asthma is a complex heterogeneous disease characterized by variable airflow obstruction, bronchial hyper-responsiveness and chronic airway inflammation. Limited therapeutic effectiveness of currently available asthma therapies warrants identification of new drug compounds. Evidence from animal studies suggests that N/OFQ modulates airway contraction and inflammation. Interestingly up regulation of the N/OFQ-NOP system reduces airway hyper-responsiveness. In contrast, inflammatory cells central to the inflammatory response in asthma may be both sources of N/OFQ and respond to NOP activation. Hence paradoxical dysregulation of the N/OFQ-NOP system may potentially play an important role in regulating airway inflammation and airway tone. To date there is no data on N/OFQ-NOP expression in the human airways. Therefore, the potential role of N/OFQ-NOP system in asthma is unknown. This review focuses on its physiological effects within airways and potential value as a novel asthma therapy.
Collapse
Affiliation(s)
- Shailendra R Singh
- Department of Cardiovascular Sciences (Pharmacology and Therapeutics Group), Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Leicester Royal Infirmary, Leicester, UK.
| | | | | | | | | |
Collapse
|
38
|
Roberto M, Gilpin NW, Siggins GR. The central amygdala and alcohol: role of γ-aminobutyric acid, glutamate, and neuropeptides. Cold Spring Harb Perspect Med 2012; 2:a012195. [PMID: 23085848 PMCID: PMC3543070 DOI: 10.1101/cshperspect.a012195] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug seeking and drug taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptative mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid-ergic (GABAergic) transmission in CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and AMPA receptors in CeA, whereas chronic alcohol up-regulates N-methyl-d-aspartate receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and anti-stress (e.g., NPY, nociceptin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.
Collapse
Affiliation(s)
- Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
39
|
Parsons MP, Burt J, Cranford A, Alberto C, Zipperlen K, Hirasawa M. Nociceptin induces hypophagia in the perifornical and lateral hypothalamic area. PLoS One 2012; 7:e45350. [PMID: 23028954 PMCID: PMC3444493 DOI: 10.1371/journal.pone.0045350] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 08/15/2012] [Indexed: 11/18/2022] Open
Abstract
Nociceptin/orphanin FQ (N/OFQ) is known to induce food intake when administered into the lateral ventricle or certain brain areas. This is somewhat contradictory to its reward-suppressing role, as food is a strong rewarding stimulus. This discrepancy may be due to the functional diversity of N/OFQ's target brain areas. N/OFQ has been shown to inhibit orexin and melanin-concentrating hormone (MCH) neurons, both of which are appetite-inducing cells. As the expression of these neurons is largely confined to the lateral hypothalamus/perifornical area (LH/PFA), we hypothesized that N/OFQ inhibits food intake by acting in this area. To test this hypothesis, we examined the effect of local N/OFQ infusion within the LH/PFA on food intake in the rat and found that N/OFQ decreased sugar pellet as well as chow intake. This effect was not seen when the injection site was outside of the LH/PFA, suggesting a site-specific effect. Next, to determine a possible cellular mechanism of N/OFQ action on food intake, whole cell patch clamp recordings were performed on rat orexin neurons. As previously reported in mice, N/OFQ induced a strong and long lasting hyperpolarization. Pharmacological study indicated that N/OFQ directly inhibited orexin neurons by activating ATP-sensitive potassium (KATP) channels. This effect was partially but significantly attenuated by the inhibitors of PI3K, PKC and PKA, suggesting that the N/OFQ signaling is mediated by these protein kinases. In summary, our results demonstrate a KATP channel-dependent N/OFQ signaling and that N/OFQ is a site-specific anorexic peptide.
Collapse
Affiliation(s)
- Matthew P. Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| | - Julia Burt
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| | - Amanda Cranford
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| | - Christian Alberto
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| | - Katrin Zipperlen
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| |
Collapse
|
40
|
Nativio P, Pascale E, Maffei A, Scaccianoce S, Passarelli F. Effect of stress on hippocampal nociceptin expression in the rat. Stress 2012; 15:378-84. [PMID: 22074385 DOI: 10.3109/10253890.2011.627071] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nociceptin/orphanin FQ (N/OFQ) peptide and its receptor are not only ubiquitously expressed in mammalian brain and spinal cord but are also abundant in limbic structures, particularly in the hippocampus. The widespread distribution of N/OFQ reflects the broad spectrum of its biological actions such as nociception, food intake, spontaneous locomotor activity, and learning and memory processes. Since the hippocampus is involved in the control of adrenocortical activity, its role in stress-related phenomena is well characterized. In male Wistar rats, we first examined the effects of acute restraint stress (120 min) on the brain immunohistochemical localization of N/OFQ. The analysis carried out on sections obtained at the onset of stress revealed enhanced expression of N/OFQ in CA1, CA3, and the dentate gyrus as well as increased plasma corticosterone concentrations. Next, we examined whether endogenous glucocorticoid hormone plays a role in the modulation of hippocampal N/OFQ expression in response to stress. To this end, rats were injected with corticosterone (1 mg/kg) or subjected to restraint stress 1 week after adrenalectomy. Two hours after corticosterone administration, plasma glucocorticoid concentrations were comparable to those observed after restraint stress, while N/OFQ expression had significantly increased in all the hippocampal subfields examined. By contrast, in adrenalectomized rats, stress did not modify protein expression. These results confirm that stress can affect N/OFQ expression and that glucocorticoids may constitute hormonal mediators of this complex interplay.
Collapse
Affiliation(s)
- Paola Nativio
- Dipartimento di Medicina Molecolare, Università La Sapienza, Roma, Italy
| | | | | | | | | |
Collapse
|
41
|
Goeldner C, Spooren W, Wichmann J, Prinssen EP. Further characterization of the prototypical nociceptin/orphanin FQ peptide receptor agonist Ro 64-6198 in rodent models of conflict anxiety and despair. Psychopharmacology (Berl) 2012; 222:203-14. [PMID: 22249359 DOI: 10.1007/s00213-012-2636-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 12/30/2011] [Indexed: 12/18/2022]
Abstract
RATIONALE Ro 64-6198, the prototypical non-peptide nociceptin/orphanin FQ peptide (NOP) receptor agonist, has potent anxiolytic-like effects in several preclinical models and species. However the effects of Ro 64-6198 on distinctive anxiety-provoking conditions related to unconditioned conflict behavior as well as its role in despair-like behavior remain to be addressed. OBJECTIVE Here we examined the effects of Ro 64-6198 on unconditioned conflict anxiety using stimuli with different salience and on regulation of autonomic reactivity and compared these to the effects of benzodiazepine receptor agonists. We also addressed the potential effects of Ro 64-6198 on despair-like behavior. MATERIALS AND METHODS Ro 64-6198 (0.1 to 10 mg/kg i.p.) and either diazepam or chlordiazepoxide were tested in the Vogel conflict punished drinking test (VCT) in Sprague Dawley rats, in the social approach-avoidance (SAA) test in Lewis rats, in the novelty-induced hypophagia (NIH) in C57BL/6J mice, and in stress-induced hyperthermia in NMRI mice, as well as in the forced swim test (FST) in Sprague Dawley rats and the tail suspension test (TST) in C57BL/6J mice. RESULTS Ro 64-6198 (0.3 to 3 mg/kg) dose-dependently produced anxiolytic-like effects in the VCT, SAA, NIH, and SIH, similar to benzodiazepine receptor agonists. Ro 64-6198 did not alter immobility time in the FST and TST. CONCLUSIONS Ro 64-6198 produced marked anxiolytic-like effects in response to a variety of mild to strong anxiogenic stimuli, whereas it did not facilitate depression-related behaviors. This data extend previous literature suggesting that NOP receptors are a viable target for the treatment of anxiety disorders.
Collapse
Affiliation(s)
- Celia Goeldner
- CNS Research, CNS Discovery, pRED, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | | | | | | |
Collapse
|
42
|
Cruz MT, Herman MA, Kallupi M, Roberto M. Nociceptin/orphanin FQ blockade of corticotropin-releasing factor-induced gamma-aminobutyric acid release in central amygdala is enhanced after chronic ethanol exposure. Biol Psychiatry 2012; 71:666-76. [PMID: 22153590 PMCID: PMC3838304 DOI: 10.1016/j.biopsych.2011.10.032] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 10/26/2011] [Accepted: 10/29/2011] [Indexed: 11/24/2022]
Abstract
BACKGROUND The central nucleus of the amygdala (CeA) mediates stress- and addiction-related processes. Corticotropin-releasing factor (CRF) and nociceptin/orphanin FQ (nociceptin) regulate ethanol intake and anxiety-like behavior. In the rat, CRF and ethanol significantly augment CeA gamma-aminobutyric acid (GABA) release, whereas nociceptin diminishes it. METHODS Using electrophysiologic techniques in an in vitro slice preparation, we investigated the interaction of nociceptin and CRF on evoked and spontaneous GABAergic transmission in CeA slices of naive and ethanol-dependent rats and the mechanistic role of protein kinase A. RESULTS In neurons from naive animals, nociceptin dose-dependently diminished basal-evoked GABA(A) receptor-mediated inhibitory postsynaptic potentials (IPSPs) by decreasing GABA release and prevented, as well as reversed, CRF-induced augmentation of IPSPs, actions that required PKA signaling. In neurons from ethanol-dependent animals, nociceptin decreased basal GABAergic transmission and blocked the CRF-induced increase in GABA release to a greater extent than in naive controls. CONCLUSIONS These data provide new evidence for an interaction between the nociceptin and CRF systems in the CeA. Nociceptin opposes CRF effects on CeA GABAergic transmission with sensitization of this effect in dependent animals. These properties of nociceptin may underlie its anti-alcohol and anxiolytic properties and identify the nociceptin receptor as a useful therapeutic target for alcoholism.
Collapse
Affiliation(s)
- Maureen T Cruz
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
43
|
2',6'-dimethylphenylalanine: a useful aromatic amino Acid surrogate for tyr or phe residue in opioid peptides. INTERNATIONAL JOURNAL OF MEDICINAL CHEMISTRY 2012; 2012:498901. [PMID: 25954528 PMCID: PMC4412257 DOI: 10.1155/2012/498901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 03/15/2012] [Accepted: 03/18/2012] [Indexed: 11/17/2022]
Abstract
Two aromatic amino acids, Tyr(1) and Phe(3) or Phe(4), are important structural elements in opioid peptides because they interact with opioid receptors. The usefulness of an artificial amino acid residue 2',6'-dimethylphenylalanine (Dmp) was investigated as an aromatic amino acid surrogate for several opioid peptides, including enkephalin, dermorphin, deltorphin, endomorphin, dynorphin A, and nociceptin peptides. In most peptides, substitutions of Phe(3) by a Dmp residue produced analogs with improved receptor-binding affinity and selectivity, while the same substitution of Phe(4) induced markedly reduced receptor affinity and selectivity. Interestingly, replacement of Tyr(1) by Dmp produced analogs with unexpectedly high affinity or produced only a slight drop in receptor affinity and bioactivity for most peptides. Thus, Dmp is also a useful surrogate for the N-terminal Tyr residue in opioid peptides despite the lack of a phenolic hydroxyl group, which is considered necessary for opioid activity. The Dmp(1)-substituted analogs are superior to 2',6'-dimethyltyrosine (Dmt)(1)-substituted analogs for high receptor selectivity since the latter generally have poor receptor selectivity. Thus, Dmp is very useful as an aromatic amino acid surrogate in opioid peptides and may be useful for developing other novel peptide mimetics with high receptor specificity.
Collapse
|
44
|
|
45
|
Matsushima A, Nishimura H, Inamine S, Uemura S, Shimohigashi Y. Capturing of the free cysteine residue in the ligand-binding site by affinity labeling of the ORL1 nociceptin receptor. Bioorg Med Chem 2011; 19:7597-602. [DOI: 10.1016/j.bmc.2011.10.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 10/07/2011] [Accepted: 10/07/2011] [Indexed: 11/25/2022]
|
46
|
Gavioli EC, Romão PRT. NOP Receptor Ligands as Potential Agents for Inflammatory and Autoimmune Diseases. JOURNAL OF AMINO ACIDS 2011; 2011:836569. [PMID: 22312472 PMCID: PMC3268226 DOI: 10.4061/2011/836569] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 08/31/2011] [Accepted: 09/24/2011] [Indexed: 12/29/2022]
Abstract
Nociceptin/orphanin FQ (N/OFQ) is a seventeen-amino acid peptide that is the endogenous ligand of a G-protein-coupled receptor (NOP). Various immune cells express the precursor protein and secrete N/OFQ as well as display binding sites for this peptide. The functional capacity of NOP receptor was demonstrated in vitro and in vivo studies by the ability of N/OFQ to induce chemotaxis of immune cells, to regulate the expression of cytokines and other inflammatory mediators, and to control cellular and humoral immunity. In this context, N/OFQ could modulate the outcome of some inflammatory diseases, such as sepsis and autoimmune pathologies by mechanisms not clearly elucidated yet. In fact, human body fluid revealed increased levels of N/OFQ under sepsis, arthritis, and Parkinson's diagnose. Preclinical studies pointed to the blockade of NOP receptor signaling as successful in treating these experimental conditions. Further preclinical and clinical studies are required to investigate the potential of NOP ligands in treating inflammatory diseases.
Collapse
Affiliation(s)
- Elaine C Gavioli
- Laboratório de Farmacologia Comportamental, Programa de Pós-graduação em Desenvolvimento e Inovação Tecnológica em Medicamentos, Departamento de Biofísica e Farmacologia, Centro de Biociências, Universidade Federal do Rio Grande do Norte, 59072-970 Natal, RN, Brazil
| | | |
Collapse
|
47
|
Lee CWS, Yan JY, Chiang YC, Hung TW, Wang HL, Chiou LC, Ho IK. Differential pharmacological actions of methadone and buprenorphine in human embryonic kidney 293 cells coexpressing human μ-opioid and opioid receptor-like 1 receptors. Neurochem Res 2011; 36:2008-21. [PMID: 21671107 PMCID: PMC3183316 DOI: 10.1007/s11064-011-0525-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2011] [Indexed: 11/26/2022]
Abstract
Methadone and buprenorphine are used in maintenance therapy for heroin addicts. In this study, we compared their effects on adenylate cyclase (AC) activity in human embryonic kidney (HEK) 293 cells stably overexpressing human μ-opioid receptor (MOR) and nociceptin/opioid receptor-like 1 receptor (ORL1) simultaneously. After acute exposure, methadone inhibited AC activity; however, buprenorphine induced compromised AC inhibition. When naloxone was introduced after 30 min incubation with methadone, the AC activity was enhanced. This was not observed in the case of buprenorphine. Enhancement of the AC activity was more significant when the incubation lasted for 4 h, and prolonged exposure to buprenorphine elevated the AC activity as well. The removal of methadone and buprenorphine by washing also obtained similar AC superactivation as that revealed by naloxone challenge. The study demonstrated that methadone and buprenorphine exert initially different yet eventually convergent adaptive changes of AC activity in cells coexpressing human MOR and ORL1 receptors.
Collapse
Affiliation(s)
- Cynthia Wei-Sheng Lee
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Jia-Ying Yan
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Yao-Chang Chiang
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Tsai-Wei Hung
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Hung-Li Wang
- Department of Physiology, Chang Gung University School of Medicine, Kwei-San, Taoyuan, 33302 Taiwan
| | - Lih-Chu Chiou
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051 Taiwan
| | - Ing-Kang Ho
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| |
Collapse
|
48
|
Bongsebandhu-Phubhakdi S, Phisonkulkasem T, Srikiatkhachorn A. Nociceptin/orphanin FQ modulates cortical activity and trigeminal nociception. Headache 2011; 51:1245-53. [PMID: 21797854 DOI: 10.1111/j.1526-4610.2011.01958.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Alterations in the levels of nociceptin/orphanin FQ (N/OFQ) have been reported in patients with primary headaches, including migraines and cluster headaches. These clinical observations suggest that N/OFQ is involved in the pathogenesis of primary headaches. OBJECTIVES The present study was conducted to determine the role of N/OFQ in the control of trigeminal nociception and cortical excitation. METHODS Cortical spreading depression (CSD) was elicited in Wistar rats by cortical application of potassium chloride, and electrocorticograms were recorded. N/OFQ was administered via an intracisternal injection. The presence of CSD-evoked trigeminal nociception was determined with Fos and transient receptor potential vanilloid 1 (TRPV1) immunoreactivity. RESULTS Nociceptin/orphanin FQ produced a biphasic effect on CSD generation, characterized by an initial attenuation followed by delayed potentiation. The amplitude of CSD waves were lower in the initial period but increased in the later period. The total number of CSD waves recorded in 1 hour was greater in the N/OFQ-treated group. Exposure to N/OFQ significantly increased the number of Fos-immunoreactive cells in the trigeminal nucleus caudalis and the number of TRPV1-immunoreactive cells in the trigeminal ganglia, indicating the enhancement of trigeminal nociception. CONCLUSION These results indicate that N/OFQ can lead to biphasic effect characterized by an initial inhibition, and delay potentiation that eventually intensify CSD-evoked trigeminal nociception.
Collapse
|
49
|
Mika J, Obara I, Przewlocka B. The role of nociceptin and dynorphin in chronic pain: implications of neuro-glial interaction. Neuropeptides 2011; 45:247-61. [PMID: 21477860 DOI: 10.1016/j.npep.2011.03.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 03/15/2011] [Accepted: 03/15/2011] [Indexed: 01/08/2023]
Abstract
Nociceptin-opioid peptide (NOP) receptor, also known as opioid receptor like-1 (ORL1), was identified following the cloning of the kappa-opioid peptide (KOP) receptor, and the characterization of these receptors revealed high homology. The endogenous ligand of NOP, nociceptin (NOC), which shares high homology to dynorphin (DYN), was discovered shortly thereafter, and since then, it has been the subject of several investigations. Despite the many advances in our understanding of the involvement of NOC and DYN systems in pain, tolerance and withdrawal, the precise function of these systems has not been fully characterized. Here, we review the recent literature concerning the distribution of the NOC and DYN systems in the central nervous system and the involvement of these systems in nociceptive transmission, especially under chronic pain conditions. We discuss the use of endogenous and exogenous ligands of NOP and KOP receptors in pain perception, as well as the potential utility of NOP ligands in clinical practice for pain management. We also discuss the modulation of opioid effects by NOC and DYN. We emphasize the important role of neuro-glial interactions in the effects of NOC and DYN, focusing on their presence in neuronal and non-neuronal cells and the changes associated with chronic pain conditions. We also present the dynamics of immune and glial regulation of neuronal functions and the importance of this regulation in the roles of NOC and DYN under conditions of neuropathic pain and in the use of drugs that alter these systems for better control of neuropathic pain.
Collapse
Affiliation(s)
- Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | | | | |
Collapse
|
50
|
Lu SX, Higgins GA, Hodgson RA, Hyde LA, Del Vecchio RA, Guthrie DH, Kazdoba T, McCool MF, Morgan CA, Bercovici A, Ho GD, Tulshian D, Parker EM, Hunter JC, Varty GB. The anxiolytic-like profile of the nociceptin receptor agonist, endo-8-[bis(2-chlorophenyl)methyl]-3-phenyl-8-azabicyclo[3.2.1]octane-3-carboxamide (SCH 655842): comparison of efficacy and side effects across rodent species. Eur J Pharmacol 2011; 661:63-71. [PMID: 21545797 DOI: 10.1016/j.ejphar.2011.04.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 03/28/2011] [Accepted: 04/14/2011] [Indexed: 11/19/2022]
Abstract
The endogenous opioid-like peptide, nociceptin, produces anxiolytic-like effects that are mediated via the nociceptin (NOP) receptor. Similarly, synthetic, non-peptide NOP agonists produce robust anxiolytic-like effects although these effects are limited by marked side effects. In the present studies, the effects of a novel NOP receptor agonist, SCH 655842, were examined in rodent models sensitive to anxiolytic drugs and tests measuring potential adverse affects. Oral administration of SCH 655842 produced robust, anxiolytic-like effects in three species, i.e., rat, guinea pig, and mouse. Specifically, SCH 655842 was effective in rat conditioned lick suppression (3-10 mg/kg) and fear-potentiated startle (3-10 mg/kg) tests, a guinea pig pup vocalization test (1-3 mg/kg), as well as in mouse Geller-Seifter (30 mg/kg) and marble burying (30 mg/kg) tests. The anxiolytic-like effect of SCH 655842 in the conditioned lick suppression test was attenuated by the NOP antagonist, J-113397. In mice, SCH 655842 reduced locomotor activity and body temperature at doses similar to the anxiolytic-like dose and these effects were absent in NOP receptor knockout mice. In rats, SCH 655842 did not produce adverse behavioral effects up to doses of 70-100 mg/kg. Pharmacokinetic studies in the rat confirmed dose-related increases in plasma and brain levels of SCH 655842 across a wide oral dose range. Taken together, SCH 655842 may represent a NOP receptor agonist with improved tolerability compared to other members of this class although further studies are necessary to establish whether this extends to higher species.
Collapse
Affiliation(s)
- Sherry X Lu
- Department of Neurobiology, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|