Top ▲

NOP receptor

Click here for help

Target not currently curated in GtoImmuPdb

Target id: 320

Nomenclature: NOP receptor

Family: Opioid receptors

Gene and Protein Information Click here for help
class A G protein-coupled receptor
Species TM AA Chromosomal Location Gene Symbol Gene Name Reference
Human 7 370 20q13.33 OPRL1 opioid related nociceptin receptor 1 66
Mouse 7 367 2 103.74 cM Oprl1 opioid receptor-like 1 75
Rat 7 367 3q43 Oprl1 opioid related nociceptin receptor 1 9,91
Previous and Unofficial Names Click here for help
N/OFQ receptor | OP4 | KOR-3 | NOCIR | kappa3-related opioid receptor | MOR-C | nociceptin receptor ORL1 | XOR1 | NOP-r | nociceptin/orphanin FQ receptor | NOPr
Database Links Click here for help
Specialist databases
GPCRdb oprx_human (Hs), oprx_mouse (Mm), oprx_rat (Rn)
Other databases
Alphafold
ChEMBL Target
Ensembl Gene
Entrez Gene
Human Protein Atlas
KEGG Gene
OMIM
Pharos
RefSeq Nucleotide
RefSeq Protein
SynPHARM
UniProtKB
Wikipedia
Selected 3D Structures Click here for help
Image of receptor 3D structure from RCSB PDB
Description:  Structure of the N/OFQ Opioid Receptor in Complex with a Peptide Mimetic
PDB Id:  4EA3
Ligand:  compound 24 [PMID: 16451050]
Resolution:  3.01Å
Species:  Human
References:  84
Natural/Endogenous Ligands Click here for help
nociceptin/orphanin FQ {Sp: Human, Mouse, Rat}
Principal endogenous agonists
nociceptin/orphanin FQ (PNOC, Q13519)  [1,8,73]

Download all structure-activity data for this target as a CSV file go icon to follow link

Agonists
Key to terms and symbols View all chemical structures Click column headers to sort
Ligand Sp. Action Value Parameter Reference
UFP-102 Peptide Hs Full agonist 10.7 pKd 15
pKd 10.7 [15]
[3H]Leu-N/OFQ Peptide Ligand is labelled Ligand is radioactive Hs Full agonist 10.2 pKd 57
pKd 10.2 (Kd 6x10-11 M) [57]
[3H]N/OFQ Peptide Ligand is labelled Ligand is radioactive Hs Full agonist 9.8 – 10.2 pKd 23,64
pKd 9.8 – 10.2 (Kd 1.5x10-10 – 6.3x10-11 M) [23,64]
[3H]Tyr14-N/OFQ Peptide Ligand is labelled Ligand is radioactive Mm Full agonist 10.0 pKd 1
pKd 10.0 [1]
[(pF)Phe4]N/OFQ-(1-13)-NH2 Peptide Hs Full agonist 10.8 – 11.3 pKi 8,33
pKi 10.8 – 11.3 [8,33]
N/OFQ-NH2 Peptide Hs Full agonist 10.4 pKi 57
pKi 10.4 [57]
PWT2-N/OFQ Peptide Hs Agonist 10.3 pKi 77
pKi 10.3 [77]
N/OFQ-(1-13)-NH2 Peptide Hs Full agonist 10.1 – 10.4 pKi 8,31,57,73
pKi 10.1 – 10.4 [8,31,57,73]
MCOPPB Small molecule or natural product Hs Full agonist 10.1 pKi 35
pKi 10.1 (Ki 8.6x10-11 M) [35]
Description: Binding affinity for hNOP
nociceptin/orphanin FQ {Sp: Human, Mouse, Rat} Peptide Hs Full agonist 9.7 – 10.4 pKi 1,8,73
pKi 9.7 – 10.4 [1,8,73]
(R)-Ro65-6570 Small molecule or natural product Hs Agonist 9.6 pKi 92
pKi 9.6 [92]
Ac-RYYRWK-NH2 Peptide Hs Partial agonist 9.1 – 10.0 pKi 23,57
pKi 9.1 – 10.0 (Ki 7.1x10-10 – 1x10-10 M) [23,57]
SCH221510 Small molecule or natural product Click here for species-specific activity table Hs Agonist 9.5 pKi 89
pKi 9.5 (Ki 3x10-10 M) [89]
Description: Radioligand binding assay
[Arg14Lys15]N/OFQ Peptide Hs Full agonist 9.5 pKi 72
pKi 9.5 [72]
Ro64-6198 Small molecule or natural product Hs Full agonist 9.4 pKi 42,93
pKi 9.4 [42,93]
nociceptin/orphanin FQ {Sp: Human, Mouse, Rat} Peptide Ligand is endogenous in the given species Mm Full agonist 9.4 pKi 23
pKi 9.4 [23]
SCH221510 Small molecule or natural product Rn Agonist 9.4 pKi 89
pKi 9.4 (Ki 4.2x10-10 M) [89]
Description: Radioligand binding assay
Ac-RYYRWK-NH2 Peptide Mm Partial agonist 9.2 pKi 23
pKi 9.2 [23]
[F/G]N/OFQ-(1-13)-NH2 Peptide Hs Partial agonist 9.2 pKi 57
pKi 9.2 [57]
cebranopadol Small molecule or natural product Click here for species-specific activity table Hs Agonist 9.1 pKi 49
pKi 9.1 (Ki 9x10-10 M) [49]
Description: Radioligand binding assay
cebranopadol Small molecule or natural product Click here for species-specific activity table Rn Agonist 9.0 pKi 49
pKi 9.0 (Ki 1x10-9 M) [49]
Description: Radioligand binding assay
Ac-RYYRIK-NH2 Peptide Hs Partial agonist 8.8 – 9.1 pKi 23,57
pKi 8.8 – 9.1 (Ki 1.5x10-9 – 7.9x10-10 M) [23,57]
AT-403 Small molecule or natural product Hs Agonist 9.0 pKi 3
pKi 9.0 (Ki 1.1x10-9 M) [3]
SCH221510 Small molecule or natural product Mm Agonist 8.9 pKi 89
pKi 8.9 (Ki 1.15x10-9 M) [89]
Description: Radioligand binding assay
Ac-RYYRIK-NH2 Peptide Mm Partial agonist 8.8 pKi 23
pKi 8.8 [23]
SCH486757 Small molecule or natural product Hs Agonist 8.3 pKi 58
pKi 8.3 (Ki 4.6x10-9 M) [58]
Description: Radioligand binding assay
BU08028 Small molecule or natural product Click here for species-specific activity table Hs Partial agonist 8.1 pKi 46
pKi 8.1 (Ki 8.46x10-9 M) [46]
SR16835 Small molecule or natural product Click here for species-specific activity table Hs Agonist 7.9 pKi 86
pKi 7.9 (Ki 1.14x10-8 M) [86]
Description: Radioligand binding assay
[(pF)Phe4]N/OFQ-(1-13)-NH2 Peptide Hs Agonist 10.1 pEC50 33
pEC50 10.1 [33]
Description: Stimulation of GTP-γ35S binding
MCOPPB Small molecule or natural product Hs Full agonist 9.4 pEC50 35
pEC50 9.4 (EC50 3.9x10-10 M) [35]
Description: Potency determined in a functional assay; [35S]GTPγS recruitment
nociceptin/orphanin FQ {Sp: Human, Mouse, Rat} Peptide Hs Agonist 8.4 – 9.8 pEC50 23
pEC50 8.4 – 9.8 [23]
UFP-112 Peptide Hs Full agonist 8.4 – 9.7 pEC50 14,78
pEC50 8.4 – 9.7 [14,78]
PWT2-N/OFQ Peptide Hs Agonist 8.8 pEC50 77
pEC50 8.8 [77]
Description: Stimulation of GTP-γ35S binding
Ac-RYYRWK-NH2 Peptide Hs Agonist 8.7 pEC50 23
pEC50 8.7 (EC50 2.1x10-9 M) [23]
SCH221510 Small molecule or natural product Click here for species-specific activity table Hs Agonist 7.9 pEC50 89
pEC50 7.9 (EC50 1.2x10-8 M) [89]
Description: Stimulation of GTP-γ35S binding
cebranopadol Small molecule or natural product Click here for species-specific activity table Hs Agonist 7.9 pEC50 49
pEC50 7.9 (EC50 1.3x10-8 M) [49]
Description: Stimulation of GTP-γ35S binding
Ro64-6198 Small molecule or natural product Hs Agonist 7.4 pEC50 93
pEC50 7.4 [93]
Description: Stimulation of GTPγ35S binding
(R)-Ro65-6570 Small molecule or natural product Hs Agonist 7.4 pEC50 92
pEC50 7.4 [92]
Description: Stimulation of GTP-γ35S binding
SR16835 Small molecule or natural product Click here for species-specific activity table Hs Agonist 7.3 pEC50 86
pEC50 7.3 (EC50 4.61x10-8 M) [86]
Description: Stimulation of GTP-γ35S binding
SCH486757 Small molecule or natural product Hs Agonist 7.1 pEC50 58
pEC50 7.1 (EC50 7.9x10-8 M) [58]
Description: Stimulation of GTP-γ35S binding
Ac-RYYRIK-NH2 Peptide Hs Agonist 5.1 pEC50 23
pEC50 5.1 [23]
Description: Stimulation of GTP-γ35S binding
View species-specific agonist tables
Agonist Comments
The above affinities are based on the use of radiolabelled N/OFQ to bind to membrane preparations from CHO cells containing the NOP receptors. This represents a high affinity binding conformation in the absence of Na+ and GTP, and low affinity values are not available. The Ki in intact cells, or in the presence of Na+ and GTP analogues can be different and multiple affinity sites have been observed.

Discrimination of full or partial agonism is very dependent on the level of receptor expression and on the assay used to monitor agonist effects. For instance [Phe1ψ(CH2-NH)Gly2]NC(1-13)NH2, the first "antagonist" reported [32], has antagonist activity in the guinea pig ileum and mouse vas deferens, but partial agonist activity in transfected cells, and full agonist activity in vivo. The identification of agonist activity in the table is largely based on the ability to stimulate GTPγS binding in cell lines expressing cloned human NOP receptors and in some instances smooth muscle bioassays such as mouse or rat vas deferens. Agents giving 85% or greater stimulation than that given by N/OFQ have been characterized as Full Agonists [23,57]. Results may be different in brain membranes. In vivo and smooth muscle bioassay results may also be different and depend upon the assay performed.

Biased Agonists
There are have been two publications discussing biased agonism for the NOP receptor [16,50]. In each case compounds with less than 100% efficacy with respect to G protein coupling appeared to be ineffective for β-arrestin coupling. Furthermore, potencies of agonists for the NOP receptor/β-arrestin interaction were systematically lower that those measured for the NOP receptor/G protein interaction in both publications.
Antagonists
Key to terms and symbols View all chemical structures Click column headers to sort
Ligand Sp. Action Value Parameter Reference
TRAP-101 Small molecule or natural product Mm Antagonist 7.8 pA2 87
pA2 7.8 [87]
Description: Antagonism of the electrically induced twitch response in mouse vas deferens stimulated by N/OFQ as agonist.
NiK-21273 Small molecule or natural product Hs Antagonist 7.4 pKB 53
pKB 7.4 (KB 4.17x10-8 M) [53]
Description: Antagonism of N/OFQ-induced calcium mobilisation in CHO cells expressing hNOP.
UFP-101 Peptide Hs Antagonist 10.2 pKi 11
pKi 10.2 [11]
LY2940094 Small molecule or natural product Hs Antagonist 10.0 pKi 25,85
pKi 10.0 (Ki 1x10-10 M) [25,85]
compound 24 [PMID: 16451050] Small molecule or natural product Ligand has a PDB structure Hs Antagonist 9.6 pKi 26
pKi 9.6 (Ki 2.4x10-10 M) [26]
SB 612111 Small molecule or natural product Ligand has a PDB structure Hs Antagonist 9.2 – 9.5 pKi 83,95
pKi 9.2 – 9.5 [83,95]
AT-076 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 8.8 pKi 97
pKi 8.8 (Ki 1.75x10-9 M) [97]
Description: Radioligand binding assay
J-113397 Small molecule or natural product Hs Antagonist 8.7 pKi 74
pKi 8.7 [74]
[Nphe1]N/OFQ-(1-13)-NH2 Peptide Hs Antagonist 8.4 pKi 11
pKi 8.4 [11]
JTC-801 Small molecule or natural product Hs Antagonist 8.1 pKi 80
pKi 8.1 [80]
peptide III-BTD Peptide Hs Antagonist 7.6 pKi 4
pKi 7.6 [4]
compound 24 [PMID: 16451050] Small molecule or natural product Ligand has a PDB structure Hs Antagonist 9.6 – 10.0 pIC50 26,29
pIC50 9.6 – 10.0 (IC50 2.7x10-10 – 1x10-10 M) [26,29]
SB 612111 Small molecule or natural product Ligand has a PDB structure Hs Antagonist 8.2 – 9.7 pIC50 83,95
pIC50 8.2 – 9.7 [83,95]
J-113397 Small molecule or natural product Hs Antagonist 8.3 pIC50 45
pIC50 8.3 [45]
View species-specific antagonist tables
Antagonist Comments
The above affinities ase based on binding to receptors in membrane preparations and represent a rough average from, in some cases, multiple studies.

So far, no inverse agonists have been reported for the NOP receptor.

More details about the pharmacological profile of the NOP receptors and the chemistry of NOP ligands can be found in review articles [10,12,63,96].
Allosteric Modulator Comments
Although no small molecules are considered direct allosteric regulators of the NOP receptor, a number of proteins such as G protein-coupled receptor kinases, β-arrestins and G proteins clearly regulate receptor functions. Furthermore, sodium and guanyl nucleotides can modify the functional NOP complex and G protein interaction. Finally, other G protein-coupled receptors (i.e. the μ opioid receptor [90]) appear to be able to form heterodimers with NOP receptors, potentailly modifying the receptor activity.
Primary Transduction Mechanisms Click here for help
Transducer Effector/Response
Gi/Go family Adenylyl cyclase inhibition
Potassium channel
Calcium channel
Other - See Comments
Comments:  NOP receptors have been shown to activate MAP kinase and phospholipase C/[Ca2+] [34].
References:  20,23,61,76,88
Secondary Transduction Mechanisms Click here for help
Transducer Effector/Response
Gi/Go family Adenylyl cyclase inhibition
References: 
Tissue Distribution Click here for help
Brain: cerebral > hippocampus, cerebellum, striatum.
Species:  Human
Technique:  Radioligand binding and in situ hybridisation.
References:  5,69
CNS: cortex, olfactory bulb, suprachiasmatic nucleus, amygdala, nucleus accumbens, thalamic nuclei, hypothalamus, hippocampus, septum, superior colliculus.
Species:  Mouse
Technique:  Radioligand binding.
References:  17-19,82
CNS
Species:  Rat
Technique:  Radioligand binding and in situ hubridisation.
References:  11,27,68,81
Tissue Distribution Comments
NOP receptors are located both pre- and post-synaptically in various areas of the CNS.

Brain: cingulate, retrosplenial, perirhinal, insular and occipital cortex, anterior and posteromedial cortical amygdaloid nuclei, basolateral amygdaloid nucleus, amygdaloid complex, posterior hippocampus, dorsal endopiriform, central medial thalamic, paraventricular, rhomboid thalamic, suprachiasmatic, ventromedial hypothalamic nuclei, mammillary complex, superficial gray layer of the superior colliculus, locus coeruleus, dorsal raphe nucleus > prefrontal, fronto-parietal, temporal, piriform cortex, dentate gyrus, anterior olfactory nucleus, olfactory tubercle, shell of nucleus accumbens, claustrum, lateral septum, laterodorsal thalamic, medial habenular, subthalamic, reuniens thalamic nuclei, subiculum, periaqueductal grey matter and pons > anterior and medial hippocampus, olfactory bulb, caudate putamen, the core of the nucleus accumbens, medial septum, ventrolateral, ventroposterolateral and mediodorsal thalamic nuclei, lateral and medial geniculate nuclei, hypothalamic area, substantia nigra, ventral tegmentum area and interpedoncular nucleus.

Like the μ opioid receptor, the NOP receptor shows very dense binding in many caudal and rostral regions, but with a notably distinct binding profile. The distinction of labelling in comparison to the classical opioid receptors is most evident in the caudate putamen, where NOP receptors are relatively low. In contrast, structures such as the suprachiasmatic nucleus have an abundant expression of NOP receptors.

Studies of the distribution of NOP receptors in humans have also been limited to autoradiography and in situ hybridisation analysis. NOP receptors in the CNS appear to have a similar distribution in rat and human. It should be noted that NOP receptors appear far sooner and in larger numbers in the developing brain than the other opioid receptors do, suggesting an important role in development [68-69].
For a review of the tissue distribution of this receptor see [65].

Spinal cord: dorsal and ventral horn.

DRG: Found on large, medium, and small dorsal root ganglia neurons.
Expression Datasets Click here for help

Show »

Log average relative transcript abundance in mouse tissues measured by qPCR from Regard, J.B., Sato, I.T., and Coughlin, S.R. (2008). Anatomical profiling of G protein-coupled receptor expression. Cell, 135(3): 561-71. [PMID:18984166] [Raw data: website]

There should be a chart of expression data here, you may need to enable JavaScript!
Functional Assays Click here for help
Measurement of musculature contraction of sections of mouse vas deferens following stimulation of the intramural nerves.
Species:  Mouse
Tissue:  Vas deferens.
Response measured:  Inhibition of electrically-evoked contractions.
References:  6,13
Measurement of adrenergic neuromuscular transmission in the rat anococcygeus muscle.
This tissue is selectively sensitive to N/OFQ and does not respond to classical opioid ligands; thus it can be considered a monoreceptor preparation.
Species:  Rat
Tissue:  Anococcygeus muscle.
Response measured:  Inhibition adrenergic motor response to electrical-field stimulation.
References:  36
Measurement of cAMP levels in CHO cells transfected with the rat NOP receptor.
Species:  Rat
Tissue:  CHO cells.
Response measured:  Inhibition of forskolin-stimulated cAMP accumulation.
References:  23
Measurement of Ca2+ channel activity in SH-SY5Y neuroblastoma cells endogenously expressing the NOP receptor.
Species:  Human
Tissue:  SH-SY5Y neuroblastoma cells.
Response measured:  Inhibition of N-type calcium current.
References:  20
Measurement of cAMP levels in BE(2)-C human neuroblastoma cells endogenously expressing the human NOP receptor.
Species:  Human
Tissue:  BE(2)-C neuroblastoma cells.
Response measured:  Inhibition of forskolin-stimulated cAMP accumulation.
References:  52
Measurement of [35S]GTPγS binding in CHO cells transfected with the mouse κ receptor.
Species:  Mouse
Tissue:  CHO cells.
Response measured:  [35S]GTPγS binding.
References:  23,88
Measurement of [35S]GTPγS binding in mouse brain preperations.
Species:  Mouse
Tissue:  Brain membranes.
Response measured:  [35S]GTPγS binding.
References:  88
Measurement of cAMP levels in CHO cells transfected with the human NOP receptor.
Species:  Human
Tissue:  CHO cells.
Response measured:  Inhibition of forskolin-stimulated cAMP accumulation.
References:  61
Physiological Functions Click here for help
Inhibition of electrically-evoked muscle contraction in the vas deferens.
Species:  Mouse
Tissue:  Vas deferens.
References:  6,13
Inhibition of electrically-evoked muscle contraction of the colon.
Species:  Rat
Tissue:  Colon.
References:  59
Inhibition of electrically-evoked muscle contraction in the vas deferens.
Species:  Rat
Tissue:  Vas deferens.
References:  7
Inhibition of glutamate release.
Species:  Rat
Tissue:  Cerebrocortical slices.
References:  70
Inhibition of acetylcholine release.
Species:  Rat
Tissue:  In vivo.
References:  40
NOP agonist-induced anxiogenic activity.
Species:  Rat
Tissue:  In vivo.
References:  24
Inhibition of noradrenaline release.
Species:  Mouse
Tissue:  Cortex slices.
References:  79
Inhibition of glutamate release.
Species:  Rat
Tissue:  Cerebrocortical slices.
References:  70
Inhibition of serotonin release.
Species:  Rat
Tissue:  Neocortex.
References:  56
Contraction of the colon.
Species:  Mouse
Tissue:  Colon.
References:  59
Nociception.
Species:  Mouse
Tissue:  In vivo.
References:  38-39,61-62
NOP agonist-induced anxiolytic activity after i.c.v. or peripheral injection.
Species:  Rat
Tissue:  In vivo.
References:  42
NOP agonist-induced anxiolytic activity after i.c.v. or peripheral injection.
Species:  Mouse
Tissue:  In vivo.
References:  41
NOP agonist-induced cardiovascular depression and water diuretic activity after either i.c.v. or intravenous administration.
Species:  Rat
Tissue:  In vivo.
References:  43-44
Inhibition of opioid antinociception.
Species:  Mouse
Tissue:  In vivo i.c.v. administration
References:  61-62,76
Antinociceptive activity.
Species:  Mouse
Tissue:  In vivo, intrathecal administration
References:  30,94
Inhibition of dopamine release.
Species:  Rat
Tissue:  In vivo, measured by microdialysis
References:  22,47,67
Physiological Functions Comments
Nociception: NOP receptor agonists attenuate opiate-mediated and stress-induced analgesia when administered i.c.v. but have antinociceptive activity when administered intrathecally. Small molecule agonists and antagonists generally have little effect on pain threshold when administered alone systemically in rodents. In chronic pain states systemic administration of small molecule NOP agonists is effective for inhibition of mechanical allodynia. In non-human primates, selective NOP agonists have antinociceptive activity. NOP receptor agonists and antagonists have many behavioural functions not discussed here, for a good review on the topic see [48].
For a review on the functional architecture of the NOP receptor, including information on splice variants, see reference [60].
Physiological Consequences of Altering Gene Expression Click here for help
NOP receptor knockout mice exhibit facilitation of long-term potentiation and learning abilities.
Species:  Mouse
Tissue: 
Technique:  Gene targeting in embryonic stem cells.
References:  51
Mice lacking the gene coding for the NOP receptor (NOP-/- mice) are viable, breed well and appear to age normally.
NOP receptor knockout mice exhibit insufficient recovery of hearing ability from the adaptation to sound exposure.
Species:  Mouse
Tissue: 
Technique:  Gene targeting in embryonic stem cells.
References:  71
NOP receptor knockout mice exhibit normal sensitivity to acute nociceptive stimulation but pronociceptive phenotype in some nociceptive tests.
Species:  Mouse
Tissue: 
Technique:  Gene targeting in embryonic stem cells.
References:  21,37
NOP receptor knockout mice exhibit an antidepressant phenotype in the forced swimming assay.
Species:  Mouse
Tissue: 
Technique:  Gene targeting in embryonic stem cells.
References:  28
NOP receptor knockout mice exhibit a better locomotor performance in the rotarod test and less sensitivity to haloperidol-induced motor depression.
Species:  Mouse
Tissue: 
Technique:  Gene targeting in embryonic stem cells.
References:  54-55
NOP receptor knockout mice exhibit attenuation of morphine tolerance and dependence.
Species:  Mouse
Tissue: 
Technique:  Gene targeting in embryonic stem cells.
References:  88
Phenotypes, Alleles and Disease Models Click here for help Mouse data from MGI

Show »

Allele Composition & genetic background Accession Phenotype Id Phenotype Reference
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0002735 abnormal chemical nociception PMID: 12814369  9155012 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0002063 abnormal learning/memory/conditioning PMID: 9707118 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae
MGI:97440  MP:0002734 abnormal mechanical nociception PMID: 16519664 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0002799 abnormal passive avoidance behavior PMID: 9707118 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0001413 abnormal response to new environment PMID: 9707118 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0001463 abnormal spatial learning PMID: 9707118 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0001968 abnormal touch/ nociception PMID: 9155012 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0001982 decreased chemically-elicited antinociception PMID: 9660760 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0003998 decreased thermal nociceptive threshold PMID: 12814369 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0003008 enhanced long term potentiation PMID: 9707118 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0009757 impaired behavioral response to morphine PMID: 11027224 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0001906 increased dopamine level PMID: 15030410 
Oprl1tm1Hta Oprl1tm1Hta/Oprl1tm1Hta
involves: 129S4/SvJae * C57BL/6J
MGI:97440  MP:0004597 increased susceptibility to noise-induced hearing loss PMID: 9155012 
Oprl1tm1Dgen Oprl1tm1Dgen/Oprl1tm1Dgen
involves: 129P2/OlaHsd * C57BL/6
MGI:97440  MP:0002169 no abnormal phenotype detected
Biologically Significant Variants Click here for help
Type:  Single nucleotide polymorphism
Species:  Human
Description:  SNP rs6010719 is associated with increased PTSD symptoms.
SNP accession: 
References:  2
Type:  Single nucleotide polymorphism
Species:  Human
Description:  SNP rs6010718 is linked with alcoholism
SNP accession: 

References

Show »

1. Adapa ID, Toll L. (1997) Relationship between binding affinity and functional activity of nociceptin/orphanin FQ. Neuropeptides, 31 (5): 403-8. [PMID:9413015]

2. Andero R, Brothers SP, Jovanovic T, Chen YT, Salah-Uddin H, Cameron M, Bannister TD, Almli L, Stevens JS, Bradley B et al.. (2013) Amygdala-dependent fear is regulated by Oprl1 in mice and humans with PTSD. Sci Transl Med, 5 (188): 188ra73. [PMID:23740899]

3. Arcuri L, Novello S, Frassineti M, Mercatelli D, Pisanò CA, Morella I, Fasano S, Journigan BV, Meyer ME, Polgar WE et al.. (2018) Anti-Parkinsonian and anti-dyskinetic profiles of two novel potent and selective nociceptin/orphanin FQ receptor agonists. Br J Pharmacol, 175 (5): 782-796. [PMID:29232769]

4. Becker JA, Wallace A, Garzon A, Ingallinella P, Bianchi E, Cortese R, Simonin F, Kieffer BL, Pessi A. (1999) Ligands for kappa-opioid and ORL1 receptors identified from a conformationally constrained peptide combinatorial library. J Biol Chem, 274 (39): 27513-22. [PMID:10488086]

5. Berthele A, Platzer S, Dworzak D, Schadrack J, Mahal B, Büttner A, Assmus HP, Wurster K, Zieglgänsberger W, Conrad B et al.. (2003) [3H]-nociceptin ligand-binding and nociceptin opioid receptor mrna expression in the human brain. Neuroscience, 121 (3): 629-40. [PMID:14568023]

6. Berzetei-Gurske IP, Schwartz RW, Toll L. (1996) Determination of activity for nociceptin in the mouse vas deferens. Eur J Pharmacol, 302 (1-3): R1-2. [PMID:8791013]

7. Bigoni R, Giuliani S, Calo' G, Rizzi A, Guerrini R, Salvadori S, Regoli D, Maggi CA. (1999) Characterization of nociceptin receptors in the periphery: in vitro and in vivo studies. Naunyn Schmiedebergs Arch Pharmacol, 359 (3): 160-7. [PMID:10208302]

8. Bigoni R, Rizzi D, Rizzi A, Camarda V, Guerrini R, Lambert DG, Hashiba E, Berger H, Salvadori S, Regoli D et al.. (2002) Pharmacological characterisation of [(pX)Phe4]nociceptin(1-13)amide analogues. 1. In vitro studies. Naunyn Schmiedebergs Arch Pharmacol, 365 (6): 442-9. [PMID:12070757]

9. Bunzow JR, Saez C, Mortrud M, Bouvier C, Williams JT, Low M, Grandy DK. (1994) Molecular cloning and tissue distribution of a putative member of the rat opioid receptor gene family that is not a mu, delta or kappa opioid receptor type. FEBS Lett, 347 (2-3): 284-8. [PMID:8034019]

10. Calo G, Guerrini R, Rizzi A, Salvadori S, Burmeister M, Kapusta DR, Lambert DG, Regoli D. (2005) UFP-101, a peptide antagonist selective for the nociceptin/orphanin FQ receptor. CNS Drug Rev, 11: 97-112. [PMID:16007234]

11. Calo G, Rizzi A, Rizzi D, Bigoni R, Guerrini R, Marzola G, Marti M, McDonald J, Morari M, Lambert DG et al.. (2002) [Nphe1,Arg14,Lys15]nociceptin-NH2, a novel potent and selective antagonist of the nociceptin/orphanin FQ receptor. Br J Pharmacol, 136 (2): 303-11. [PMID:12010780]

12. Calo' G, Bigoni R, Rizzi A, Guerrini R, Salvadori S, Regoli D. (2000) Nociceptin/orphanin FQ receptor ligands. Peptides, 21 (7): 935-47. [PMID:10998527]

13. Calò G, Rizzi A, Bogoni G, Neugebauer V, Salvadori S, Guerrini R, Bianchi C, Regoli D. (1996) The mouse vas deferens: a pharmacological preparation sensitive to nociceptin. Eur J Pharmacol, 311 (1): R3-5. [PMID:8884244]

14. Camarda V, Fischetti C, Anzellotti N, Molinari P, Ambrosio C, Kostenis E, Regoli D, Trapella C, Guerrini R, Severo S et al.. (2009) Pharmacological profile of NOP receptors coupled with calcium signaling via the chimeric protein G alpha qi5. Naunyn Schmiedebergs Arch Pharmacol, 379 (6): 599-607. [PMID:19183962]

15. Carrà G, Rizzi A, Guerrini R, Barnes TA, McDonald J, Hebbes CP, Mela F, Kenigs VA, Marzola G, Rizzi D et al.. (2005) [(pF)Phe4,Arg14,Lys15]N/OFQ-NH2 (UFP-102), a highly potent and selective agonist of the nociceptin/orphanin FQ receptor. J Pharmacol Exp Ther, 312 (3): 1114-23. [PMID:15509719]

16. Chang SD, Mascarella SW, Spangler SM, Gurevich VV, Navarro HA, Carroll FI, Bruchas MR. (2015) Quantitative Signaling and Structure-Activity Analyses Demonstrate Functional Selectivity at the Nociceptin/Orphanin FQ Opioid Receptor. Mol Pharmacol, 88 (3): 502-11. [PMID:26134494]

17. Clarke S, Chen Z, Hsu MS, Hill RG, Pintar JE, Kitchen I. (2003) Nociceptin/orphanin FQ knockout mice display up-regulation of the opioid receptor-like 1 receptor and alterations in opioid receptor expression in the brain. Neuroscience, 117 (1): 157-68. [PMID:12605902]

18. Clarke S, Chen Z, Hsu MS, Pintar J, Hill R, Kitchen I. (2001) Quantitative autoradiographic mapping of the ORL1, mu-, delta- and kappa-receptors in the brains of knockout mice lacking the ORL1 receptor gene. Brain Res, 906: 13-24. [PMID:11430857]

19. Clarke S, Czyzyk T, Ansonoff M, Nitsche JF, Hsu MS, Nilsson L, Larsson K, Borsodi A, Toth G, Hill R, Kitchen I, Pintar JE. (2002) Autoradiography of opioid and ORL1 ligands in opioid receptor triple knockout mice. Eur J Neurosci, 16: 1705-1712. [PMID:12431223]

20. Connor M, Yeo A, Henderson G. (1996) The effect of nociceptin on Ca2+ channel current and intracellular Ca2+ in the SH-SY5Y human neuroblastoma cell line. Br J Pharmacol, 118 (2): 205-7. [PMID:8735615]

21. Depner UB, Reinscheid RK, Takeshima H, Brune K, Zeilhofer HU. (2003) Normal sensitivity to acute pain, but increased inflammatory hyperalgesia in mice lacking the nociceptin precursor polypeptide or the nociceptin receptor. Eur J Neurosci, 17 (11): 2381-7. [PMID:12814369]

22. Di Giannuario A, Pieretti S, Catalani A, Loizzo A. (1999) Orphanin FQ reduces morphine-induced dopamine release in the nucleus accumbens: a microdialysis study in rats. Neurosci Lett, 272 (3): 183-6. [PMID:10505611]

23. Dooley CT, Spaeth CG, Berzetei-Gurske IP, Craymer K, Adapa ID, Brandt SR, Houghten RA, Toll L. (1997) Binding and in vitro activities of peptides with high affinity for the nociceptin/orphanin FQ receptor, ORL1. J Pharmacol Exp Ther, 283 (2): 735-41. [PMID:9353393]

24. Fernandez F, Misilmeri MA, Felger JC, Devine DP. (2004) Nociceptin/orphanin FQ increases anxiety-related behavior and circulating levels of corticosterone during neophobic tests of anxiety. Neuropsychopharmacology, 29 (1): 59-71. [PMID:14532912]

25. Ferrari F, Rizzo S, Ruzza C, Calo G. (2020) Detailed In Vitro Pharmacological Characterization of the Clinically Viable Nociceptin/Orphanin FQ Peptide Receptor Antagonist BTRX-246040. J Pharmacol Exp Ther, 373 (1): 34-43. [PMID:31937563]

26. Fischetti C, Camarda V, Rizzi A, Pelà M, Trapella C, Guerrini R, McDonald J, Lambert DG, Salvadori S, Regoli D et al.. (2009) Pharmacological characterization of the nociceptin/orphanin FQ receptor non peptide antagonist Compound 24. Eur J Pharmacol, 614 (1-3): 50-7. [PMID:19445927]

27. Florin S, Meunier J, Costentin J. (2000) Autoradiographic localization of [3H]nociceptin binding sites in the rat brain. Brain Res, 880 (1-2): 11-6. [PMID:11032985]

28. Gavioli EC, Marzola G, Guerrini R, Bertorelli R, Zucchini S, De Lima TC, Rae GA, Salvadori S, Regoli D, Calo G. (2003) Blockade of nociceptin/orphanin FQ-NOP receptor signalling produces antidepressant-like effects: pharmacological and genetic evidences from the mouse forced swimming test. Eur J Neurosci, 17 (9): 1987-90. [PMID:12752799]

29. Goto Y, Arai-Otsuki S, Tachibana Y, Ichikawa D, Ozaki S, Takahashi H, Iwasawa Y, Okamoto O, Okuda S, Ohta H et al.. (2006) Identification of a novel spiropiperidine opioid receptor-like 1 antagonist class by a focused library approach featuring 3D-pharmacophore similarity. J Med Chem, 49 (3): 847-9. [PMID:16451050]

30. Grisel JE, Mogil JS, Belknap JK, Grandy DK. (1996) Orphanin FQ acts as a supraspinal, but not a spinal, anti-opioid peptide. Neuroreport, 7 (13): 2125-9. [PMID:8930972]

31. Guerrini R, Calo G, Rizzi A, Bianchi C, Lazarus LH, Salvadori S, Temussi PA, Regoli D. (1997) Address and message sequences for the nociceptin receptor: a structure-activity study of nociceptin-(1-13)-peptide amide. J Med Chem, 40 (12): 1789-93. [PMID:9191955]

32. Guerrini R, Calo G, Rizzi A, Bigoni R, Bianchi C, Salvadori S, Regoli D. (1998) A new selective antagonist of the nociceptin receptor. Br J Pharmacol, 123 (2): 163-5. [PMID:9489602]

33. Guerrini R, Caló G, Lambert DG, Carrá G, Arduin M, Barnes TA, McDonald J, Rizzi D, Trapella C, Marzola E et al.. (2005) N- and C-terminal modifications of nociceptin/orphanin FQ generate highly potent NOP receptor ligands. J Med Chem, 48 (5): 1421-7. [PMID:15743186]

34. Hawes BE, Graziano MP, Lambert DG. (2000) Cellular actions of nociceptin: transduction mechanisms. Peptides, 21: 961-967. [PMID:10998529]

35. Hayashi S, Hirao A, Imai A, Nakamura H, Murata Y, Ohashi K, Nakata E. (2009) Novel non-peptide nociceptin/orphanin FQ receptor agonist, 1-[1-(1-Methylcyclooctyl)-4-piperidinyl]-2-[(3R)-3-piperidinyl]-1H-benzimidazole: design, synthesis, and structure-activity relationship of oral receptor occupancy in the brain for orally potent antianxiety drug. J Med Chem, 52 (3): 610-25. [PMID:19125610]

36. Ho M, Corbett AD, McKnight AT. (2000) Characterization of the ORL(1) receptor on adrenergic nerves in the rat anococcygeus muscle. Br J Pharmacol, 131 (2): 349-55. [PMID:10991930]

37. Inoue M, Kawashima T, Takeshima H, Calo G, Inoue A, Nakata Y, Ueda H. (2003) In vivo pain-inhibitory role of nociceptin/orphanin FQ in spinal cord. J Pharmacol Exp Ther, 305 (2): 495-501. [PMID:12606680]

38. Inoue M, Kobayashi M, Kozaki S, Zimmer A, Ueda H. (1998) Nociceptin/orphanin FQ-induced nociceptive responses through substance P release from peripheral nerve endings in mice. Proc Natl Acad Sci USA, 95 (18): 10949-53. [PMID:9724810]

39. Inoue M, Shimohira I, Yoshida A, Zimmer A, Takeshima H, Sakurada T, Ueda H. (1999) Dose-related opposite modulation by nociceptin/orphanin FQ of substance P nociception in the nociceptors and spinal cord. J Pharmacol Exp Ther, 291 (1): 308-13. [PMID:10490918]

40. Itoh K, Konya H, Takai E, Masuda H, Nagai K. (1999) Modification of acetylcholine release by nociceptin in conscious rat striatum. Brain Res, 845 (2): 242-5. [PMID:10536205]

41. Jenck F, Moreau JL, Martin JR, Kilpatrick GJ, Reinscheid RK, Monsma Jr FJ, Nothacker HP, Civelli O. (1997) Orphanin FQ acts as an anxiolytic to attenuate behavioral responses to stress. Proc Natl Acad Sci USA, 94 (26): 14854-8. [PMID:9405703]

42. Jenck F, Wichmann J, Dautzenberg FM, Moreau JL, Ouagazzal AM, Martin JR, Lundstrom K, Cesura AM, Poli SM, Roever S et al.. (2000) A synthetic agonist at the orphanin FQ/nociceptin receptor ORL1: anxiolytic profile in the rat. Proc Natl Acad Sci USA, 97 (9): 4938-43. [PMID:10758169]

43. Kapusta DR, Burmeister MA, Calo' G, Guerrini R, Gottlieb HB, Kenigs VA. (2005) Functional selectivity of nociceptin/orphanin FQ peptide receptor partial agonists on cardiovascular and renal function. J Pharmacol Exp Ther, 314 (2): 643-51. [PMID:15855356]

44. Kapusta DR, Kenigs VA. (1999) Cardiovascular and renal responses produced by central orphanin FQ/nociceptin occur independent of renal nerves. Am J Physiol, 277 (4): R987-95. [PMID:10516236]

45. Kawamoto H, Ozaki S, Itoh Y, Miyaji M, Arai S, Nakashima H, Kato T, Ohta H, Iwasawa Y. (1999) Discovery of the first potent and selective small molecule opioid receptor-like (ORL1) antagonist: 1-[(3R,4R)-1-cyclooctylmethyl-3- hydroxymethyl-4-piperidyl]-3-ethyl-1, 3-dihydro-2H-benzimidazol-2-one (J-113397). J Med Chem, 42 (25): 5061-3. [PMID:10602690]

46. Khroyan 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 (3): 952-61. [PMID:21177476]

47. Konya H, Masuda H, Itoh K, Nagai K, Kakishita E, Matsuoka A. (1998) Modification of dopamine release by nociceptin in conscious rat striatum. Brain Res, 788 (1-2): 341-4. [PMID:9555088]

48. Lambert DG. (2008) The nociceptin/orphanin FQ receptor: a target with broad therapeutic potential. Nat Rev Drug Discov, 7 (8): 694-710. [PMID:18670432]

49. Linz K, Christoph T, Tzschentke TM, Koch T, Schiene K, Gautrois M, Schröder W, Kögel BY, Beier H, Englberger W et al.. (2014) Cebranopadol: a novel potent analgesic nociceptin/orphanin FQ peptide and opioid receptor agonist. J Pharmacol Exp Ther, 349 (3): 535-48. [PMID:24713140]

50. Malfacini D, Ambrosio C, Gro' MC, Sbraccia M, Trapella C, Guerrini R, Bonora M, Pinton P, Costa T, Calo' G. (2015) Pharmacological Profile of Nociceptin/Orphanin FQ Receptors Interacting with G-Proteins and β-Arrestins 2. PLoS ONE, 10 (8): e0132865. [PMID:26248189]

51. Manabe T, Noda Y, Mamiya T, Katagiri H, Houtani T, Nishi M, Noda T, Takahashi T, Sugimoto T, Nabeshima T et al.. (1998) Facilitation of long-term potentiation and memory in mice lacking nociceptin receptors. Nature, 394 (6693): 577-81. [PMID:9707118]

52. Mandyam CD, Altememi GF, Standifer KM. (2000) beta-Funaltrexamine inactivates ORL1 receptors in BE(2)-C human neuroblastoma cells. Eur J Pharmacol, 402 (1-2): R1-37. [PMID:10940375]

53. Marti M, Mela F, Budri M, Volta M, Malfacini D, Molinari S, Zaveri NT, Ronzoni S, Petrillo P, Calò G et al.. (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 (4): 863-79. [PMID:22994368]

54. Marti M, Mela F, Fantin M, Zucchini S, Brown JM, Witta J, Di Benedetto M, Buzas B, Reinscheid RK, Salvadori S et al.. (2005) Blockade of nociceptin/orphanin FQ transmission attenuates symptoms and neurodegeneration associated with Parkinson's disease. J Neurosci, 25 (42): 9591-601. [PMID:16237164]

55. Marti M, Mela F, Veronesi C, Guerrini R, Salvadori S, Federici M, Mercuri NB, Rizzi A, Franchi G, Beani L et al.. (2004) Blockade of nociceptin/orphanin FQ receptor signaling in rat substantia nigra pars reticulata stimulates nigrostriatal dopaminergic transmission and motor behavior. J Neurosci, 24 (30): 6659-66. [PMID:15282268]

56. Marti M, Stocchi S, Paganini F, Mela F, De Risi C, Calo' G, Guerrini R, Barnes TA, Lambert DG, Beani L et al.. (2003) Pharmacological profiles of presynaptic nociceptin/orphanin FQ receptors modulating 5-hydroxytryptamine and noradrenaline release in the rat neocortex. Br J Pharmacol, 138 (1): 91-8. [PMID:12522077]

57. McDonald J, Barnes TA, Okawa H, Williams J, Calo' G, Rowbotham DJ, Lambert DG. (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 (1): 61-70. [PMID:12967935]

58. McLeod RL, Tulshian DB, Bolser DC, Varty GB, Baptista M, Fernandez X, Parra LE, Zimmer JC, Erickson CH, Ho GD et al.. (2010) Pharmacological profile of the NOP agonist and cough suppressing agent SCH 486757 (8-[Bis(2-Chlorophenyl)Methyl]-3-(2-Pyrimidinyl)-8-Azabicyclo[3.2.1]Octan-3-Ol) in preclinical models. Eur J Pharmacol, 630 (1-3): 112-20. [PMID:20006596]

59. Menzies JR, Glen T, Davies MR, Paterson SJ, Corbett AD. (1999) In vitro agonist effects of nociceptin and [Phe(1)psi(CH(2)-NH)Gly(2)]nociceptin(1-13)NH(2) in the mouse and rat colon and the mouse vas deferens. Eur J Pharmacol, 385 (2-3): 217-23. [PMID:10607879]

60. Meunier J, Mouledous L, Topham CM. (2000) The nociceptin (ORL1) receptor: molecular cloning and functional architecture. Peptides, 21 (7): 893-900. [PMID:10998522]

61. Meunier J-C, Mollereau C, Toll L, Suaudeau C, Moisand C, Alvinerie P, Butour JL, Guillemot JC, Ferrara P, Monsarrat B et al.. (1995) Isolation and structure of the endogenous agonist of opioid receptor-like ORL-1 receptor. Nature, 377: 532-535. [PMID:7566152]

62. Mogil JS, Grisel JE, Zhangs G, Belknap JK, Grandy DK. (1996) Functional antagonism of mu-, delta- and kappa-opioid antinociception by orphanin FQ. Neurosci Lett, 214 (2-3): 131-4. [PMID:8878101]

63. Mogil JS, Pasternak GW. (2001) The molecular and behavioral pharmacology of the orphanin FQ/nociceptin peptide and receptor family. Pharmacol Rev, 53: 381-415. [PMID:11546835]

64. Mollereau C, Moisand C, Butour JL, Parmentier M, Meunier JC. (1996) Replacement of Gln280 by His in TM6 of the human ORL1 receptor increases affinity but reduces intrinsic activity of opioids. FEBS Lett, 395 (1): 17-21. [PMID:8849681]

65. Mollereau C, Mouledous L. (2000) Tissue distribution of the opioid receptor-like (ORL1) receptor. Peptides, 21 (7): 907-17. [PMID:10998524]

66. Mollereau C, Parmentier M, Mailleux P, Butour JL, Moisand C, Chalon P, Caput D, Vassart G, Meunier JC. (1994) ORL1, a novel member of the opioid receptor family. Cloning, functional expression and localization. FEBS Lett, 341 (1): 33-8. [PMID:8137918]

67. Murphy NP, Ly HT, Maidment NT. (1996) Intracerebroventricular orphanin FQ/nociceptin suppresses dopamine release in the nucleus accumbens of anaesthetized rats. Neuroscience, 75 (1): 1-4. [PMID:8923516]

68. Neal CR, Mansour A, Reinscheid R, Nothacker HP, Civelli O, Akil H, Watson SJ. (1999) Opioid receptor-like (ORL1) receptor distribution in the rat central nervous system: comparison of ORL1 receptor mRNA expression with125I-[14Tyr]-orphanin FQ binding. J Comp Neurol, 412: 563-605. [PMID:10464356]

69. Neal Jr CR, Akil H, Watson Jr SJ. (2001) Expression of orphanin FQ and the opioid receptor-like (ORL1) receptor in the developing human and rat brain. J Chem Neuroanat, 22 (4): 219-49. [PMID:11719021]

70. Nicol B, Lambert DG, Rowbotham DJ, Smart D, McKnight AT. (1996) Nociceptin induced inhibition of K+ evoked glutamate release from rat cerebrocortical slices. Br J Pharmacol, 119 (6): 1081-3. [PMID:8937708]

71. Nishi M, Houtani T, Noda Y, Mamiya T, Sato K, Doi T, Kuno J, Takeshima H, Nukada T, Nabeshima T et al.. (1997) Unrestrained nociceptive response and disregulation of hearing ability in mice lacking the nociceptin/orphaninFQ receptor. EMBO J, 16 (8): 1858-64. [PMID:9155012]

72. Okada K, Sujaku T, Chuman Y, Nakashima R, Nose T, Costa T, Yamada Y, Yokoyama M, Nagahisa A, Shimohigashi Y. (2000) Highly potent nociceptin analog containing the Arg-Lys triple repeat. Biochem Biophys Res Commun, 278: 493-498. [PMID:11097863]

73. Okawa H, Nicol B, Bigoni R, Hirst RA, Calo G, Guerrini R, Rowbotham DJ, Smart D, McKnight AT, Lambert DG. (1999) Comparison of the effects of [Phe1psi(CH2-NH)Gly2]nociceptin(1-13)NH2 in rat brain, rat vas deferens and CHO cells expressing recombinant human nociceptin receptors. Br J Pharmacol, 127 (1): 123-30. [PMID:10369464]

74. Ozaki S, Kawamoto H, Itoh Y, Miyaji M, Azuma T, Ichikawa D, Nambu H, Iguchi T, Iwasawa Y, Ohta H. (2000) In vitro and in vivo pharmacological characterization of J-113397, a potent and selective non-peptidyl ORL1 receptor antagonist. Eur J Pharmacol, 402 (1-2): 45-53. [PMID:10940356]

75. Pan YX, Cheng J, Xu J, Rossi G, Jacobson E, Ryan-Moro J, Brooks AI, Dean GE, Standifer KM, Pasternak GW. (1995) Cloning and functional characterization through antisense mapping of a kappa 3-related opioid receptor. Mol Pharmacol, 47 (6): 1180-8. [PMID:7603458]

76. Reinscheid RK, Nothacker HP, Bourson A, Ardati A, Henningsen RA, Bunzow JR, Grandy DK, Langen H, Monsma Jr FJ, Civelli O. (1995) Orphanin FQ: a neuropeptide that activates an opioidlike G protein-coupled receptor. Science, 270 (5237): 792-4. [PMID:7481766]

77. Rizzi A, Malfacini D, Cerlesi MC, Ruzza C, Marzola E, Bird MF, Rowbotham DJ, Salvadori S, Guerrini R, Lambert DG et al.. (2014) In vitro and in vivo pharmacological characterization of nociceptin/orphanin FQ tetrabranched derivatives. Br J Pharmacol, 171 (17): 4138-53. [PMID:24903280]

78. Rizzi A, Spagnolo B, Wainford RD, Fischetti C, Guerrini R, Marzola G, Baldisserotto A, Salvadori S, Regoli D, Kapusta DR et al.. (2007) 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 (6): 1240-51. [PMID:17532097]

79. Schlicker E, Werthwein S, Kathmann M, Bauer U. (1998) Nociceptin inhibits noradrenaline release in the mouse brain cortex via presynaptic ORL1 receptors. Naunyn Schmiedebergs Arch Pharmacol, 358 (4): 418-22. [PMID:9826063]

80. Shinkai H, Ito T, Iida T, Kitao Y, Yamada H, Uchida I. (2000) 4-Aminoquinolines: novel nociceptin antagonists with analgesic activity. J Med Chem, 43 (24): 4667-77. [PMID:11101358]

81. Sim LJ, Xiao R, Childers SR. (1996) Identification of opioid receptor-like (ORL1) peptide-stimulated [35S]GTP gamma S binding in rat brain. Neuroreport, 7 (3): 729-33. [PMID:8733732]

82. Slowe SJ, Clarke S, Lena I, Goody RJ, Lattanzi R, Negri L, Simonin F, Matthes HW, Filliol D, Kieffer BL, Kitchen I. (2001) Autoradiographic mapping of the opioid receptor-like 1 (ORL1) receptor in the brains of mu-, delta- or kappa-opioid receptor knockout mice. Neuroscience, 106: 469-480. [PMID:11591451]

83. Spagnolo B, Carrà G, Fantin M, Fischetti C, Hebbes C, McDonald J, Barnes TA, Rizzi A, Trapella C, Fanton G et al.. (2007) 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 vitro studies. J Pharmacol Exp Ther, 321 (3): 961-7. [PMID:17329552]

84. Thompson AA, Liu W, Chun E, Katritch V, Wu H, Vardy E, Huang XP, Trapella C, Guerrini R, Calo G et al.. (2012) Structure of the nociceptin/orphanin FQ receptor in complex with a peptide mimetic. Nature, 485 (7398): 395-9. [PMID:22596163]

85. Toledo MA, Pedregal C, Lafuente C, Diaz N, Martinez-Grau MA, Jiménez A, Benito A, Torrado A, Mateos C, Joshi EM et al.. (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 (8): 3418-29. [PMID:24678969]

86. Toll 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 (3): 954-64. [PMID:19773529]

87. Trapella C, Guerrini R, Piccagli L, Calo' G, Carra' G, Spagnolo B, Rubini S, Fanton G, Hebbes C, McDonald J et al.. (2006) Identification of an achiral analogue of J-113397 as potent nociceptin/orphanin FQ receptor antagonist. Bioorg Med Chem, 14 (3): 692-704. [PMID:16202610]

88. Ueda H, Inoue M, Takeshima H, Iwasawa Y. (2000) Enhanced spinal nociceptin receptor expression develops morphine tolerance and dependence. J Neurosci, 20 (20): 7640-7. [PMID:11027224]

89. Varty GB, Lu SX, Morgan CA, Cohen-Williams ME, Hodgson RA, Smith-Torhan A, Zhang H, Fawzi AB, Graziano MP, Ho GD et al.. (2008) The anxiolytic-like effects of the novel, orally active nociceptin opioid receptor agonist 8-[bis(2-methylphenyl)methyl]-3-phenyl-8-azabicyclo[3.2.1]octan-3-ol (SCH 221510). J Pharmacol Exp Ther, 326 (2): 672-82. [PMID:18492950]

90. Wang HL, Hsu CY, Huang PC, Kuo YL, Li AH, Yeh TH, Tso AS, Chen YL. (2005) Heterodimerization of opioid receptor-like 1 and mu-opioid receptors impairs the potency of micro receptor agonist. J Neurochem, 92 (6): 1285-94. [PMID:15748148]

91. Wang JB, Johnson PS, Imai Y, Persico AM, Ozenberger BA, Eppler CM, Uhl GR. (1994) cDNA cloning of an orphan opiate receptor gene family member and its splice variant. FEBS Lett, 348 (1): 75-9. [PMID:8026588]

92. Wichmann J, Adam G, Röver S, Cesura AM, Dautzenberg FM, Jenck F. (1999) 8-acenaphthen-1-yl-1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-one derivatives as orphanin FQ receptor agonists. Bioorg Med Chem Lett, 9 (16): 2343-8. [PMID:10476866]

93. Wichmann J, Adam G, Röver S, Hennig M, Scalone M, Cesura AM, Dautzenberg FM, Jenck F. (2000) Synthesis of (1S,3aS)-8-(2,3,3a,4,5, 6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4. 5]decan-4-one, a potent and selective orphanin FQ (OFQ) receptor agonist with anxiolytic-like properties. Eur J Med Chem, 35 (9): 839-51. [PMID:11006485]

94. Xu XJ, Hao JX, Wiesenfeld-Hallin Z. (1996) Nociceptin or antinociceptin: potent spinal antinociceptive effect of orphanin FQ/nociceptin in the rat. Neuroreport, 7 (13): 2092-4. [PMID:8930965]

95. Zaratin PF, Petrone G, Sbacchi M, Garnier M, Fossati C, Petrillo P, Ronzoni S, Giardina GA, Scheideler MA. (2004) Modification of nociception and morphine tolerance by the selective opiate receptor-like orphan receptor antagonist (-)-cis-1-methyl-7-[[4-(2,6-dichlorophenyl)piperidin-1-yl]methyl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-ol (SB-612111). J Pharmacol Exp Ther, 308 (2): 454-61. [PMID:14593080]

96. Zaveri N. (2003) Peptide and nonpeptide ligands for the nociceptin/orphanin FQ receptor ORL1: research tools and potential therapeutic agents. Life Sci, 73: 663-678. [PMID:12801588]

97. Zaveri NT, Journigan VB, Polgar WE. (2015) Discovery of the first small-molecule opioid pan antagonist with nanomolar affinity at mu, delta, kappa, and nociceptin opioid receptors. ACS Chem Neurosci, 6 (4): 646-57. [PMID:25635572]

Contributors

Show »

How to cite this page