Skip to main content

NOP Receptor Ligands and Parkinson’s Disease

  • Chapter
  • First Online:
The Nociceptin/Orphanin FQ Peptide Receptor

Part of the book series: Handbook of Experimental Pharmacology ((HEP,volume 254))

Abstract

Nociceptin/Orphanin FQ (N/OFQ) and its NOP receptor are highly expressed in motor areas of the rodent, nonhuman, and human primate brain, such as primary motor cortex, thalamus, globus pallidus, striatum, and substantia nigra. Endogenous N/OFQ negatively regulates motor behavior and dopamine transmission through NOP receptors expressed by dopaminergic neurons of the substantia nigra compacta. Consistent with the existence of an N/OFQ tone over dopaminergic transmission, blockade of NOP receptor antagonists increases striatal dopamine release. In this chapter, we will review the evidence linking the N/OFQ-NOP receptor system to Parkinson’s disease (PD). We will first discuss data showing that the central N/OFQ-NOP receptor system undergoes plastic changes in different basal ganglia nuclei following dopamine depletion. Then we will show that NOP receptor antagonists relieve motor deficits in different rodent and nonhuman primate models of PD. Mechanistically, NOP receptor blockade in substantia nigra reticulata results in rebalancing of the inhibitory GABAergic and excitatory glutamatergic inputs impinging on nigro-thalamic GABAergic neurons, leading to thalamic disinhibition. We will also present data showing that, in addition to motor symptoms, N/OFQ also plays a role in the parkinsonian neurodegeneration. In fact, NOP receptor antagonists possess neuroprotective/neurorescue properties in in vitro and in vivo models of PD.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  • Albin RL, Young AB, Penney JB (1989) The functional anatomy of basal ganglia disorders. Trends Neurosci 12:366–375

    Article  CAS  Google Scholar 

  • Alexander GE, Crutcher MD, DeLong MR (1990) Basal ganglia-thalamocortical circuits: parallel substrates for motor, oculomotor, “prefrontal” and “limbic” functions. Prog Brain Res 85:119–146

    Article  CAS  Google Scholar 

  • Arcuri L, Viaro R, Bido S, Longo F, Calcagno M, Fernagut PO, Zaveri NT, Calo G, Bezard E, Morari M (2016) Genetic and pharmacological evidence that endogenous nociceptin/orphanin FQ contributes to dopamine cell loss in Parkinson’s disease. Neurobiol Dis 89:55–64

    Article  CAS  Google Scholar 

  • Arcuri L, Mercatelli D, Morari M (2017) Parkinson’s disease: no NOP, new hope. Oncotarget 8:8995–8996

    Article  Google Scholar 

  • Arcuri L, Novello S, Frassineti M, Mercatelli D, Pisano CA, Morella I, Fasano S, Journigan BV, Meyer ME, Polgar WE, Brambilla R, Zaveri NT, Morari M (2018) Anti-Parkinsonian and anti-dyskinetic profiles of two novel potent and selective nociceptin/orphanin FQ receptor agonists. Br J Pharmacol 175:782–796

    Article  CAS  Google Scholar 

  • Baik JH, Picetti R, Saiardi A, Thiriet G, Dierich A, Depaulis A, Le Meur M, Borrelli E (1995) Parkinsonian-like locomotor impairment in mice lacking dopamine D2 receptors. Nature 377:424–428

    Article  CAS  Google Scholar 

  • Bastias-Candia S, di Benedetto M, D’Addario C, Candeletti S, Romualdi P (2015) Combined exposure to agriculture pesticides, paraquat and maneb, induces alterations in the N/OFQ-NOPr and PDYN/KOPr systems in rats: relevance to sporadic Parkinson’s disease. Environ Toxicol 30:656–663

    Article  CAS  Google Scholar 

  • Brown JM, Gouty S, Iyer V, Rosenberger J, Cox BM (2006) Differential protection against MPTP or methamphetamine toxicity in dopamine neurons by deletion of ppN/OFQ expression. J Neurochem 98:495–505

    Article  CAS  Google Scholar 

  • Cenci MA, Lee CS, Bjorklund A (1998) L-DOPA-induced dyskinesia in the rat is associated with striatal overexpression of prodynorphin- and glutamic acid decarboxylase mRNA. Eur J Neurosci 10:2694–2706

    Article  CAS  Google Scholar 

  • Collins LM, Dal Bo G, Calcagno M, Monzon-Sandoval J, Sullivan AM, Gutierrez H, Morari M, O’Keeffe GW (2015) Nociceptin/orphanin FQ inhibits the survival and axon growth of midbrain dopaminergic neurons through a p38-MAPK dependent mechanism. Mol Neurobiol 53:7284–7297

    Article  CAS  Google Scholar 

  • Cuenda A, Rousseau S (2007) p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim Biophys Acta 1773:1358–1375

    Article  CAS  Google Scholar 

  • di Benedetto M, Cavina C, D’Addario C, Leoni G, Candeletti S, Cox BM, Romualdi P (2009) Alterations of N/OFQ and NOP receptor gene expression in the substantia nigra and caudate putamen of MPP+ and 6-OHDA lesioned rats. Neuropharmacology 56:761–767

    Article  CAS  Google Scholar 

  • Gavioli EC, Calo G (2013) Nociceptin/orphanin FQ receptor antagonists as innovative antidepressant drugs. Pharmacol Ther 140:10–25

    Article  CAS  Google Scholar 

  • Gouty S, Brown JM, Rosenberger J, Cox BM (2010) MPTP treatment increases expression of pre-pro-nociceptin/orphanin FQ mRNA in a subset of substantia nigra reticulata neurons. Neuroscience 169:269–278

    Article  CAS  Google Scholar 

  • Karunakaran S, Saeed U, Mishra M, Valli RK, Joshi SD, Meka DP, Seth P, Ravindranath V (2008) Selective activation of p38 mitogen-activated protein kinase in dopaminergic neurons of substantia nigra leads to nuclear translocation of p53 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice. J Neurosci 28:12500–12509

    Article  CAS  Google Scholar 

  • Khan MS, Boileau I, Kolla N, Mizrahi R (2018) A systematic review of the role of the nociceptin receptor system in stress, cognition, and reward: relevance to schizophrenia. Transl Psychiatry 8:38

    Article  CAS  Google Scholar 

  • Laudenbach V, Calo G, Guerrini R, Lamboley G, Benoist JF, Evrard P, Gressens P (2001) Nociceptin/orphanin FQ exacerbates excitotoxic white-matter lesions in the murine neonatal brain. J Clin Invest 107:457–466

    Article  CAS  Google Scholar 

  • Mabrouk OS, Marti M, Morari M (2010) Endogenous nociceptin/orphanin FQ (N/OFQ) contributes to haloperidol-induced changes of nigral amino acid transmission and parkinsonism: a combined microdialysis and behavioral study in naive and nociceptin/orphanin FQ receptor knockout mice. Neuroscience 166:40–48

    Article  CAS  Google Scholar 

  • Malinowska B, Godlewski G, Schlicker E (2002) Function of nociceptin and opioid OP4 receptors in the regulation of the cardiovascular system. J Physiol Pharmacol 53:301–324

    PubMed  CAS  Google Scholar 

  • Mallimo EM, Kusnecov AW (2013) The role of orphanin FQ/nociceptin in neuroplasticity: relationship to stress, anxiety and neuroinflammation. Front Cell Neurosci 7:173

    Article  CAS  Google Scholar 

  • Marti M, Guerrini R, Beani L, Bianchi C, Morari M (2002) Nociceptin/orphanin FQ receptors modulate glutamate extracellular levels in the substantia nigra pars reticulata. A microdialysis study in the awake freely moving rat. Neuroscience 112:153–160

    Article  CAS  Google Scholar 

  • Marti M, Mela F, Guerrini R, Calo G, Bianchi C, Morari M (2004a) Blockade of nociceptin/orphanin FQ transmission in rat substantia nigra reverses haloperidol-induced akinesia and normalizes nigral glutamate release. J Neurochem 91:1501–1504

    Article  CAS  Google Scholar 

  • Marti M, Mela F, Veronesi C, Guerrini R, Salvadori S, Federici M, Mercuri NB, Rizzi A, Franchi G, Beani L, Bianchi C, Morari M (2004b) Blockade of nociceptin/orphanin FQ receptor signaling in rat substantia nigra pars reticulata stimulates nigrostriatal dopaminergic transmission and motor behavior. J Neurosci 24:6659–6666

    Article  CAS  Google Scholar 

  • Marti M, Mela F, Fantin M, Zucchini S, Brown JM, Witta J, di Benedetto M, Buzas B, Reinscheid RK, Salvadori S, Guerrini R, Romualdi P, Candeletti S, Simonato M, Cox BM, Morari M (2005) Blockade of nociceptin/orphanin FQ transmission attenuates symptoms and neurodegeneration associated with Parkinson’s disease. J Neurosci 25:9591–9601

    Article  CAS  Google Scholar 

  • Marti M, Trapella C, Viaro R, Morari M (2007) The nociceptin/orphanin FQ receptor antagonist J-113397 and L-DOPA additively attenuate experimental parkinsonism through overinhibition of the nigrothalamic pathway. J Neurosci 27:1297–1307

    Article  CAS  Google Scholar 

  • Marti M, Trapella C, Morari M (2008) The novel nociceptin/orphanin FQ receptor antagonist Trap-101 alleviates experimental parkinsonism through inhibition of the nigro-thalamic pathway: positive interaction with L-DOPA. J Neurochem 107:1683–1696

    Article  CAS  Google Scholar 

  • Marti M, Sarubbo S, Latini F, Cavallo M, Eleopra R, Biguzzi S, Lettieri C, Conti C, Simonato M, Zucchini S, Quatrale R, Sensi M, Candeletti S, Romualdi P, Morari M (2010) Brain interstitial nociceptin/orphanin FQ levels are elevated in Parkinson’s disease. Mov Disord 25:1723–1732

    Article  Google Scholar 

  • Marti M, Rodi D, Li Q, Guerrini R, Fasano S, Morella I, Tozzi A, Brambilla R, Calabresi P, Simonato M, Bezard E, Morari M (2012) Nociceptin/orphanin FQ receptor agonists attenuate L-DOPA-induced dyskinesias. J Neurosci 32:16106–16119

    Article  CAS  Google Scholar 

  • Marti M, Mela F, Budri M, Volta M, Malfacini D, Molinari S, Zaveri NT, Ronzoni S, Petrillo P, Calo G, Morari M (2013) Acute and chronic antiparkinsonian effects of the novel nociceptin/orphanin FQ receptor antagonist NiK-21273 in comparison with SB-612111. Br J Pharmacol 168:863–879

    Article  CAS  Google Scholar 

  • Meredith GE, Rademacher DJ (2011) MPTP mouse models of Parkinson’s disease: an update. J Parkinson’s Dis 1:19–33

    CAS  Google Scholar 

  • Morari M, O’Connor WT, Darvelid M, Ungerstedt U, Bianchi C, Fuxe K (1996) Functional neuroanatomy of the nigrostriatal and striatonigral pathways as studied with dual probe microdialysis in the awake rat – I. Effects of perfusion with tetrodotoxin and low-calcium medium. Neuroscience 72:79–87

    Article  CAS  Google Scholar 

  • Nolan YM, Sullivan AM, Toulouse A (2013) Parkinson’s disease in the nuclear age of neuroinflammation. Trends Mol Med 19:187–196

    Article  CAS  Google Scholar 

  • Norton CS, Neal CR, Kumar S, Akil H, Watson SJ (2002) Nociceptin/orphanin FQ and opioid receptor-like receptor mRNA expression in dopamine systems. J Comp Neurol 444:358–368

    Article  CAS  Google Scholar 

  • Picetti R, Saiardi A, Abdel Samad T, Bozzi Y, Baik JH, Borrelli E (1997) Dopamine D2 receptors in signal transduction and behavior. Crit Rev Neurobiol 11:121–142

    Article  CAS  Google Scholar 

  • Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE, Lang AE (2017) Parkinson disease. Nat Rev Dis Prim 3:17013

    Article  Google Scholar 

  • Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos ES, Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Johnson WG, Lazzarini AM, Duvoisin RC, di Iorio G, Golbe LI, Nussbaum RL (1997) Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 276:2045–2047

    Article  CAS  Google Scholar 

  • Post A, Smart TS, Krikke-Workel J, Dawson GR, Harmer CJ, Browning M, Jackson K, Kakar R, Mohs R, Statnick M, Wafford K, McCarthy A, Barth V, Witkin JM (2016) A selective nociceptin receptor antagonist to treat depression: evidence from preclinical and clinical studies. Neuropsychopharmacology 41:1803–1812

    Article  CAS  Google Scholar 

  • Redrobe JP, Calo G, Guerrini R, Regoli D, Quirion R (2000) [Nphe(1)]-Nociceptin (1-13)-NH(2), a nociceptin receptor antagonist, reverses nociceptin-induced spatial memory impairments in the Morris water maze task in rats. Br J Pharmacol 131:1379–1384

    Article  CAS  Google Scholar 

  • Scatton B, Claustre Y, Cudennec A, Oblin A, Perrault G, Sanger DJ, Schoemaker H (1997) Amisulpride: from animal pharmacology to therapeutic action. Int Clin Psychopharmacol 12(Suppl 2):S29–S36

    Article  Google Scholar 

  • Schoemaker H, Claustre Y, Fage D, Rouquier L, Chergui K, Curet O, Oblin A, Gonon F, Carter C, Benavides J, Scatton B (1997) Neurochemical characteristics of amisulpride, an atypical dopamine D2/D3 receptor antagonist with both presynaptic and limbic selectivity. J Pharmacol Exp Ther 280:83–97

    PubMed  CAS  Google Scholar 

  • Schwarting RK, Huston JP (1996a) The unilateral 6-hydroxydopamine lesion model in behavioral brain research. Analysis of functional deficits, recovery and treatments. Prog Neurobiol 50:275–331

    Article  CAS  Google Scholar 

  • Schwarting RK, Huston JP (1996b) Unilateral 6-hydroxydopamine lesions of meso-striatal dopamine neurons and their physiological sequelae. Prog Neurobiol 49:215–266

    Article  CAS  Google Scholar 

  • Sedelis M, Hofele K, Auburger GW, Morgan S, Huston JP, Schwarting RK (2000) MPTP susceptibility in the mouse: behavioral, neurochemical, and histological analysis of gender and strain differences. Behav Genet 30:171–182

    Article  CAS  Google Scholar 

  • Serra PA, Sciola L, Delogu MR, Spano A, Monaco G, Miele E, Rocchitta G, Miele M, Migheli R, Desole MS (2002) The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine induces apoptosis in mouse nigrostriatal glia. Relevance to nigral neuronal death and striatal neurochemical changes. J Biol Chem 277:34451–34461

    Article  CAS  Google Scholar 

  • Sibaev A, Fichna J, Saur D, Yuece B, Timmermans JP, Storr M (2015) Nociceptin effect on intestinal motility depends on opioid-receptor like-1 receptors and nitric oxide synthase co-localization. World J Gastrointest Pharmacol Ther 6:73–83

    Article  Google Scholar 

  • Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M (1997) Alpha-synuclein in Lewy bodies. Nature 388:839–840

    Article  CAS  Google Scholar 

  • Toledo MA, Pedregal C, Lafuente C, Diaz N, Martinez-Grau MA, Jimenez A, Benito A, Torrado A, Mateos C, Joshi EM, Kahl SD, Rash KS, Mudra DR, Barth VN, Shaw DB, McKinzie D, Witkin JM, Statnick MA (2014) Discovery of a novel series of orally active nociceptin/orphanin FQ (NOP) receptor antagonists based on a dihydrospiro(piperidine-4,7′-thieno[2,3-c]pyran) scaffold. J Med Chem 57:3418–3429

    Article  CAS  Google Scholar 

  • Toll L, Bruchas MR, Calo G, Cox BM, Zaveri NT (2016) Nociceptin/orphanin FQ receptor structure, signaling, ligands, functions, and interactions with opioid systems. Pharmacol Rev 68:419–457

    Article  Google Scholar 

  • Usiello A, Baik JH, Rouge-Pont F, Picetti R, Dierich A, LeMeur M, Piazza PV, Borrelli E (2000) Distinct functions of the two isoforms of dopamine D2 receptors. Nature 408:199–203

    Article  CAS  Google Scholar 

  • Viaro R, Sanchez-Pernaute R, Marti M, Trapella C, Isacson O, Morari M (2008) Nociceptin/orphanin FQ receptor blockade attenuates MPTP-induced parkinsonism. Neurobiol Dis 30:430–438

    Article  CAS  Google Scholar 

  • Viaro R, Calcagno M, Marti M, Borrelli E, Morari M (2013) Pharmacological and genetic evidence for pre- and postsynaptic D2 receptor involvement in motor responses to nociceptin/orphanin FQ receptor ligands. Neuropharmacology 72:126–138

    Article  CAS  Google Scholar 

  • Visanji NP, de Bie RM, Johnston TH, McCreary AC, Brotchie JM, Fox SH (2008) The nociceptin/orphanin FQ (NOP) receptor antagonist J-113397 enhances the effects of levodopa in the MPTP-lesioned nonhuman primate model of Parkinson’s disease. Mov Disord 23:1922–1925

    Article  Google Scholar 

  • Volta M, Mabrouk OS, Bido S, Marti M, Morari M (2010a) Further evidence for an involvement of nociceptin/orphanin FQ in the pathophysiology of Parkinson’s disease: a behavioral and neurochemical study in reserpinized mice. J Neurochem 115:1543–1555

    Article  CAS  Google Scholar 

  • Volta M, Marti M, McDonald J, Molinari S, Camarda V, Pela M, Trapella C, Morari M (2010b) Pharmacological profile and antiparkinsonian properties of the novel nociceptin/orphanin FQ receptor antagonist 1-[1-cyclooctylmethyl-5-(1-hydroxy-1-methyl-ethyl)-1,2,3,6-tetrahydro-pyri din-4-yl]-3-ethyl-1,3-dihydro-benzoimidazol-2-one (GF-4). Peptides 31:1194–1204

    Article  CAS  Google Scholar 

  • Volta M, Viaro R, Trapella C, Marti M, Morari M (2011) Dopamine-nociceptin/orphanin FQ interactions in the substantia nigra reticulata of hemiparkinsonian rats: involvement of D2/D3 receptors and impact on nigro-thalamic neurons and motor activity. Exp Neurol 228:126–137

    Article  CAS  Google Scholar 

  • Zarubin T, Han J (2005) Activation and signaling of the p38 MAP kinase pathway. Cell Res 15:11–18

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michele Morari .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Mercatelli, D., Pisanò, C.A., Novello, S., Morari, M. (2018). NOP Receptor Ligands and Parkinson’s Disease. In: Ko, MC., Caló, G. (eds) The Nociceptin/Orphanin FQ Peptide Receptor. Handbook of Experimental Pharmacology, vol 254. Springer, Cham. https://doi.org/10.1007/164_2018_199

Download citation

Publish with us

Policies and ethics