Show simple item record

[Dmt 1 ]N/OFQ(1–13)‐NH 2 : a potent nociceptin/orphanin FQ and opioid receptor universal agonist

dc.contributor.authorMolinari, Sen_US
dc.contributor.authorCamarda, Ven_US
dc.contributor.authorRizzi, Aen_US
dc.contributor.authorMarzola, Gen_US
dc.contributor.authorSalvadori, Sen_US
dc.contributor.authorMarzola, Een_US
dc.contributor.authorMolinari, Pen_US
dc.contributor.authorMcDonald, Jen_US
dc.contributor.authorKo, MCen_US
dc.contributor.authorLambert, DGen_US
dc.contributor.authorCalo', Gen_US
dc.contributor.authorGuerrini, Ren_US
dc.date.accessioned2013-01-03T19:42:09Z
dc.date.available2014-03-03T15:09:25Zen_US
dc.date.issued2013-01en_US
dc.identifier.citationMolinari, S; Camarda, V; Rizzi, A; Marzola, G; Salvadori, S; Marzola, E; Molinari, P; McDonald, J; Ko, MC; Lambert, DG; Calo', G; Guerrini, R (2013). " [Dmt 1 ]N/OFQ(1–13)‐NH 2 : a potent nociceptin/orphanin FQ and opioid receptor universal agonist." British Journal of Pharmacology (1): 151-162. <http://hdl.handle.net/2027.42/95271>en_US
dc.identifier.issn0007-1188en_US
dc.identifier.issn1476-5381en_US
dc.identifier.urihttps://hdl.handle.net/2027.42/95271
dc.publisherWiley Periodicals, Inc.en_US
dc.subject.otherGuinea Pig Ileumen_US
dc.subject.otherSpinal Corden_US
dc.subject.otherTail Withdrawal Assayen_US
dc.subject.otherMonkeyen_US
dc.subject.other[Dmt 1 ]N/OFQ(1–13)‐NH 2en_US
dc.subject.otherOpioid Receptorsen_US
dc.subject.otherCalcium Mobilizationen_US
dc.subject.otherReceptor and [ 35 S]‐GTPγS Bindingen_US
dc.subject.otherNOP Receptoren_US
dc.title[Dmt 1 ]N/OFQ(1–13)‐NH 2 : a potent nociceptin/orphanin FQ and opioid receptor universal agonisten_US
dc.typeArticleen_US
dc.rights.robotsIndexNoFollowen_US
dc.subject.hlbsecondlevelPharmacy and Pharmacologyen_US
dc.subject.hlbtoplevelHealth Sciencesen_US
dc.description.peerreviewedPeer Revieweden_US
dc.identifier.pmid22827708en_US
dc.description.bitstreamurlhttp://deepblue.lib.umich.edu/bitstream/2027.42/95271/1/bph2115.pdf
dc.identifier.doi10.1111/j.1476-5381.2012.02115.xen_US
dc.identifier.sourceBritish Journal of Pharmacologyen_US
dc.identifier.citedreferenceMustazza C, Bastanzio G ( 2011 ). Development of nociceptin receptor (NOP) agonists and antagonists. Med Res Rev 31: 605 – 648.en_US
dc.identifier.citedreferenceChampion HC, Bivalacqua TJ, Wang R, Hellstrom WJ, Kadowitz PJ ( 1998 ). [Tyr1]‐nociceptin and nociceptin have similar naloxone‐insensitive erectile activity in the cat. J Androl 19: 747 – 753.en_US
dc.identifier.citedreferenceFischetti C, Camarda V, Rizzi A, Pela′ M, Trapella C, Guerrini R et al. ( 2009 ). Pharmacological characterization of the nociceptin/orphanin FQ non peptide antagonist Compound 24. Eur J Pharmacol 614: 50 – 57.en_US
dc.identifier.citedreferenceGuerrini R, Calo G, Rizzi A, Bianchi C, Lazarus LH, Salvadori S et al. ( 1997 ). Address and message sequences for the nociceptin receptor: a structure‐activity study of nociceptin‐(1‐13)‐peptide amide. J Med Chem 40: 1789 – 1793.en_US
dc.identifier.citedreferenceHu E, Calo G, Guerrini R, Ko M ( 2009 ). Long lasting antinociceptive spinal effects in primates of the novel nociceptin/orphanin FQ receptor agonist UFP‐112. Pain 148: 107 – 113.en_US
dc.identifier.citedreferenceKhroyan TV, Zaveri NT, Polgar WE, Orduna J, Olsen C, Jiang F et al. ( 2007 ). SR 16435 [1‐(1‐(bicyclo[3.3.1]nonan‐9‐yl)piperidin‐4‐yl)indolin‐2‐one], a novel mixed nociceptin/orphanin FQ/mu‐opioid receptor partial agonist: analgesic and rewarding properties in mice. J Pharmacol Exp Ther 320: 934 – 943.en_US
dc.identifier.citedreferenceKhroyan TV, Polgar WE, Cami‐Kobeci G, Husbands SM, Zaveri NT, Toll L ( 2011 ). The first universal opioid ligand, (2S)‐2‐[(5R,6R,7R,14S)‐N‐cyclopropylmethyl‐4,5‐epoxy‐6,14‐ethano‐3‐hydroxy ‐6‐methoxymorphinan‐7‐yl]‐3,3‐dimethylpentan‐2‐ol (BU08028): characterization of the in vitro profile and in vivo behavioral effects in mouse models of acute pain and cocaine‐induced reward. J Pharmacol Exp Ther 336: 952 – 961.en_US
dc.identifier.citedreferenceKilkenny C, Browne W, Cuthill IC, Emerson M, Altman DG ( 2010 ). NC3Rs Reporting Guidelines Working Group. Br J Pharmacol 160: 1577 – 1579.en_US
dc.identifier.citedreferenceKleczkowska P, Kosson P, Ballet S, Van den Eynde I, Tsuda Y, Tourwe D et al. ( 2010 ). PK20, a new opioid‐neurotensin hybrid peptide that exhibits central and peripheral antinociceptive effects. Mol Pain 6: 86.en_US
dc.identifier.citedreferenceKo MC, Naughton NN ( 2009 ). Antinociceptive effects of nociceptin/orphanin FQ administered intrathecally in monkeys. J Pain 10: 509 – 516.en_US
dc.identifier.citedreferenceKo MC, Song MS, Edwards T, Lee H, Naughton NN ( 2004 ). The role of central mu opioid receptors in opioid‐induced itch in primates. J Pharmacol Exp Ther 310: 169 – 176.en_US
dc.identifier.citedreferenceKo MC, Wei H, Woods JH, Kennedy RT ( 2006 ). Effects of intrathecally administered nociceptin/orphanin FQ in monkeys: behavioral and Mass Spectrometric Studies. J Pharmacol Exp Ther 318: 1257 – 1264.en_US
dc.identifier.citedreferenceKo MC, Woods JH, Fantegrossi WE, Galuska CM, Wichmann J, Prinssen EP ( 2009 ). Behavioral effects of a synthetic agonist selective for nociceptin/orphanin FQ peptide receptors in monkeys. Neuropsychopharmacology 34: 2088 – 2096.en_US
dc.identifier.citedreferenceKress HG, Simpson KH, Marchettini P, Ver Donck A, Varrassi G ( 2009 ). Intrathecal therapy: what has changed with the introduction of ziconotide. Pain Pract 9: 338 – 347.en_US
dc.identifier.citedreferenceLambert DG ( 2008 ). The nociceptin/orphanin FQ receptor: a target with broad therapeutic potential. Nat Rev Drug Discov 7: 694 – 710.en_US
dc.identifier.citedreferenceMcDonald J, Barnes TA, Okawa H, Williams J, Calo G, Rowbotham DJ et al. ( 2003 ). Partial agonist behaviour depends upon the level of nociceptin/orphanin FQ receptor expression: studies using the ecdysone‐inducible mammalian expression system. Br J Pharmacol 140: 61 – 70.en_US
dc.identifier.citedreferenceMcGrath J, Drummond G, McLachlan E, Kilkenny C, Wainwright C ( 2010 ). Guidelines for reporting experiments involving animals: the ARRIVE guidelines. Br J Pharmacol 160: 1573 – 1576.en_US
dc.identifier.citedreferenceMalmberg AB, Yaksh TL ( 1992 ). Isobolographic and dose‐response analyses of the interaction between intrathecal mu and delta agonists: effects of naltrindole and its benzofuran analog (NTB). J Pharmacol Exp Ther 263: 264 – 275.en_US
dc.identifier.citedreferenceMeunier JC, Mollereau C, Toll L, Suaudeau C, Moisand C, Alvinerie P et al. ( 1995 ). Isolation and structure of the endogenous agonist of opioid receptor‐like ORL1 receptor. Nature 377: 532 – 535.en_US
dc.identifier.citedreferenceNarita M, Mizoguchi H, Oji DE, Dun NJ, Hwang BH, Nagase H et al. ( 1999a ). Identification of the G‐protein‐coupled ORL1 receptor in the mouse spinal cord by [35S]‐GTPgammaS binding and immunohistochemistry. Br J Pharmacol 128: 1300 – 1306.en_US
dc.identifier.citedreferenceNarita M, Mizoguchi H, Sora I, Uhl GR, Tseng LF ( 1999b ). Absence of G‐protein activation by mu‐opioid receptor agonists in the spinal cord of mu‐opioid receptor knockout mice. Br J Pharmacol 126: 451 – 456.en_US
dc.identifier.citedreferenceNazzaro C, Rizzi A, Salvadori S, Guerrini R, Regoli D, Zeilhofer HU et al. ( 2007 ). UFP‐101 antagonizes the spinal antinociceptive effects of nociceptin/orphanin FQ: behavioral and electrophysiological studies in mice. Peptides 28: 663 – 669.en_US
dc.identifier.citedreferenceReinscheid RK, Nothacker HP, Bourson A, Ardati A, Henningsen RA, Bunzow JR et al. ( 1995 ). Orphanin FQ: a neuropeptide that activates an opioidlike G protein‐coupled receptor. Science 270: 792 – 794.en_US
dc.identifier.citedreferenceRizzi A, Gavioli EC, Marzola G, Spagnolo B, Zucchini S, Ciccocioppo R et al. ( 2007a ). Pharmacological Characterization of the Nociceptin/Orphanin FQ Receptor Antagonist SB‐612111 [(‐)‐cis‐1‐Methyl‐7‐[[4‐(2,6‐dichlorophenyl)piperidin‐1‐yl]methyl]‐6,7,8,9‐tetrahydro‐5H benzocyclohepten‐5‐ol]: In vivo Studies. J Pharmacol Exp Ther 321: 968 – 974.en_US
dc.identifier.citedreferenceRizzi A, Spagnolo B, Wainford RD, Fischetti C, Guerrini R, Marzola G et al. ( 2007b ). In vitro and in vivo studies on UFP‐112, a novel potent and long lasting agonist selective for the nociceptin/orphanin FQ receptor. Peptides 28: 1240 – 1251.en_US
dc.identifier.citedreferenceSalvadori S, Attila M, Balboni G, Bianchi C, Bryant SD, Crescenzi O et al. ( 1995 ). Delta opioidmimetic antagonists: prototypes for designing a new generation of ultraselective opioid peptides. Mol Med 1: 678 – 689.en_US
dc.identifier.citedreferenceSchiller PW ( 2010 ). Bi‐ or multifunctional opioid peptide drugs. Life Sci 86: 598 – 603.en_US
dc.identifier.citedreferenceSim LJ, Selley DE, Childers SR ( 1995 ). In vitro autoradiography of receptor‐activated G proteins in rat brain by agonist‐stimulated guanylyl 5′‐[gamma‐[35S]thio]‐triphosphate binding. Proc Natl Acad Sci USA 92: 7242 – 7246.en_US
dc.identifier.citedreferenceSmith HS, Deer TR, Staats PS, Singh V, Sehgal N, Cordner H ( 2008 ). Intrathecal drug delivery. Pain Physician 11: S89 – S104.en_US
dc.identifier.citedreferenceSpagnolo B, Calo G, Polgar WE, Jiang F, Olsen CM, Berzetei‐Gurske I et al. ( 2008 ). Activities of mixed NOP and mu‐opioid receptor ligands. Br J Pharmacol 153: 609 – 619.en_US
dc.identifier.citedreferenceToll L, Khroyan TV, Polgar WE, Jiang F, Olsen C, Zaveri NT ( 2009 ). Comparison of the antinociceptive and antirewarding profiles of novel bifunctional nociceptin receptor/mu‐opioid receptor ligands: implications for therapeutic applications. J Pharmacol Exp Ther 331: 954 – 964.en_US
dc.identifier.citedreferenceVarani K, Rizzi A, Calo G, Bigoni R, Toth G, Guerrini R et al. ( 1999 ). Pharmacology of [Tyr1]nociceptin analogs: receptor binding and bioassay studies. Naunyn Schmiedebergs Arch Pharmacol 360: 270 – 277.en_US
dc.identifier.citedreferenceZeilhofer HU, Calo G ( 2003 ). Nociceptin/orphanin FQ and its receptor–potential targets for pain therapy? J Pharmacol Exp Ther 306: 423 – 429.en_US
dc.identifier.citedreferenceAlbrecht E, Samovilova NN, Oswald S, Baeger I, Berger H ( 1998 ). Nociceptin (orphanin FQ): high‐affinity and high‐capacity binding site coupled to low‐potency stimulation of guanylyl‐5′‐O‐(gamma‐thio)‐triphosphate binding in rat brain membranes. J Pharmacol Exp Ther 286: 896 – 902.en_US
dc.identifier.citedreferenceAlexander SP, Mathie A, Peters JA ( 2011 ). Guide to Receptors and Channels (GRAC), 5th edition. Br J Pharmacol 164 ( Suppl. 1 ): S1 – 324.en_US
dc.identifier.citedreferenceBigoni R, Giuliani S, Calo G, Rizzi A, Guerrini R, Salvadori S et al. ( 1999 ). Characterization of nociceptin receptors in the periphery: in vitro and in vivo studies. Naunyn Schmiedebergs Arch Pharmacol 359: 160 – 167.en_US
dc.identifier.citedreferenceBigoni R, Calo G, Rizzi A, Guerrini R, De Risi C, Hashimoto Y et al. ( 2000 ). In vitro characterization of J‐113397, a non‐peptide nociceptin/orphanin FQ receptor antagonist. Naunyn Schmiedebergs Arch Pharmacol 361: 565 – 568.en_US
dc.identifier.citedreferenceBryant SD, Jinsmaa Y, Salvadori S, Okada Y, Lazarus LH ( 2003 ). Dmt and opioid peptides: a potent alliance. Biopolymers 71: 86 – 102.en_US
dc.identifier.citedreferenceCalo G, Rizzi A, Bogoni G, Neugebauer V, Salvadori S, Guerrini R et al. ( 1996 ). The mouse vas deferens: a pharmacological preparation sensitive to nociceptin. Eur J Pharmacol 311: R3 – R5.en_US
dc.identifier.citedreferenceCalo G, Rizzi A, Bodin M, Neugebauer W, Salvadori S, Guerrini R et al. ( 1997 ). Pharmacological characterization of nociceptin receptor: an in vitro study. Can J Physiol Pharmacol 75: 713 – 718.en_US
dc.identifier.citedreferenceCamarda V, Calo G ( 2013 ). Chimeric G‐proteins in fluorimetric calcium assays: experience with opioid receptors. Methods Mol Biol 937: 293 – 306.en_US
dc.identifier.citedreferenceCamarda V, Fischetti C, Anzellotti N, Molinari P, Ambrosio C, Kostenis E et al. ( 2009 ). Pharmacological profile of NOP receptors coupled with calcium signaling via the chimeric protein Gα qi5. Naunyn Schmiedebergs Arch Pharmacol 379: 599 – 607.en_US
dc.identifier.citedreferenceChampion HC, Kadowitz PJ ( 1997 ). [Tyr1]‐nociceptin, a novel nociceptin analog, decreases systemic arterial pressure by a naloxone‐insensitive mechanism in the rat. Biochem Biophys Res Commun 234: 309 – 312.en_US
dc.owningcollnameInterdisciplinary and Peer-Reviewed


Files in this item

Show simple item record

Remediation of Harmful Language

The University of Michigan Library aims to describe library materials in a way that respects the people and communities who create, use, and are represented in our collections. Report harmful or offensive language in catalog records, finding aids, or elsewhere in our collections anonymously through our metadata feedback form. More information at Remediation of Harmful Language.

Accessibility

If you are unable to use this file in its current format, please select the Contact Us link and we can modify it to make it more accessible to you.