Korean J Physiol Pharmacol.  2019 Nov;23(6):427-448. 10.4196/kjpp.2019.23.6.427.

Role of nociceptin/orphanin FQ and nociceptin opioid peptide receptor in depression and antidepressant effects of nociceptin opioid peptide receptor antagonists

Affiliations
  • 1Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Korea. kyungho@korea.ac.kr

Abstract

Nociceptin/orphanin FQ (N/OFQ) and its receptor, nociceptin opioid peptide (NOP) receptor, are localized in brain areas implicated in depression including the amygdala, bed nucleus of the stria terminalis, habenula, and monoaminergic nuclei in the brain stem. N/OFQ inhibits neuronal excitability of monoaminergic neurons and monoamine release from their terminals by activation of G protein-coupled inwardly rectifying K⁺ channels and inhibition of voltage sensitive calcium channels, respectively. Therefore, NOP receptor antagonists have been proposed as a potential antidepressant. Indeed, mounting evidence shows that NOP receptor antagonists have antidepressant-like effects in various preclinical animal models of depression, and recent clinical studies again confirmed the idea that blockade of NOP receptor signaling could provide a novel strategy for the treatment of depression. In this review, we describe the pharmacological effects of N/OFQ in relation to depression and explore the possible mechanism of NOP receptor antagonists as potential antidepressants.

Keyword

Antidepressive agents; Depression; Neuropeptide; Receptors, drug

MeSH Terms

Amygdala
Antidepressive Agents
Brain
Brain Stem
Calcium Channels
Depression*
Habenula
Models, Animal
Neurons
Neuropeptides
Opioid Peptides*
Receptors, Drug
Septal Nuclei
Antidepressive Agents
Calcium Channels
Neuropeptides
Opioid Peptides
Receptors, Drug

Figure

  • Fig. 1 Electrophysiological effects of nociceptin/orphanin FQ (N/OFQ) on ion channels in neuronal cell body and axon terminals. Nociceptin opioid peptide (NOP) receptor activation by N/OFQ stimulates G protein-coupled inwardly rectifying K+ channels (GIRK) and voltage sensitive M-type K+ channels, but inhibits voltage sensitive transient outward A-type K+ channels in neuronal cell body. Presynaptic NOP receptor activation in axon terminals inhibits voltage sensitive calcium channels and delayed rectifier K+ channels. N/OFQ inhibits neuronal excitability and neurotransmitter release from axon terminals by activation of GIRK and inhibition of voltage sensitive calcium channels, respectively. For example, release of norepinephrine, dopamine, 5-hydroxytryptamine (serotonin), acetylcholine, and glutamate from their terminals are known to be inhibited by N/OFQ. This scheme is modified using Motifolio Scientific Illustration Toolkits for Presentations and Publications (www.motifolio.com).

  • Fig. 2 Inhibitory effects of nociceptin/orphanin FQ (N/OFQ) on noradrenergic neurotransmission. Nociceptin opioid peptide (NOP) receptor is distributed on norepinephrine (NE) neurons in the locus coeruleus (LC) and their terminals in the basolateral amygdala (BLA) and prefrontal cortex (PFC). N/OFQ inhibits activity of noradrenergic LC neurons and leads to diminish NE release in the BLA and PFC. There is also evidence that N/OFQ reduces NE release by acting on NOP receptor at axon terminals in the BLA and PFC, but the NE release in the BLA and PFC following NOP receptor activation is regulated in a rather different manner. Intraperitoneal injection of NOP receptor antagonist J-113397 increases NE release and the simultaneous infusion of N/OFQ into the BLA blocks the effects of J-113397 indicating that NOP receptor in the BLA appears to exert tonic inhibitory effects on NE release. On the contrary, local injection of NOP receptor agonist NCNH2 into the LC decreases NE release in the PFC and this effect is partially restored by co-administration of NOP receptor antagonist [Nphe1]NC(1–13)NH2 and NCNH2 into the LC. Interestingly, injection of [Nphe1]NC(1–13)NH2 into the LC is without effect on NE release in the PFC per se. Thus, NOP in the LC seems not to exert tonic inhibition of NE release in the PFC. In vitro studies using brain slice and synaptosome demonstrate that N/OFQ also diminishes the release of NE in the frontal cortex. This scheme is modified using Motifolio Scientific Illustration Toolkits for Presentations and Publications (www.motifolio.com).

  • Fig. 3 Inhibition of dopamine (DA) release by nociceptin/orphanin FQ (N/OFQ) in the ventral tegmental area (VTA). N/OFQ inhibits DA release in the VTA and its terminals via both direct and indirect hyperpolarization of DA neurons. N/OFQ activates NOP receptors localized on DA neurons directly followed by a decrease in its neuronal activity. GABAergic terminals in the VTA are largely apposed by enkephalinergic nerve terminals and release of GABA is often presynaptically inhibited by enkephalin. N/OFQ is known to block released enkephalin (Enk) via anti-opioid effects, and thus reduced enkephalinergic tone could increase GABA release in the VTA. Therefore, DA neuronal activity in the VTA is suppressed by direct inhibitory effect of N/OFQ along with enhanced GABAergic neurotransmission indirectly. δ, delta-opioid receptors; GABAA, γ-aminobutyric acid A receptors. This scheme is modified using Motifolio Scientific Illustration Toolkits for Presentations and Publications (www.motifolio.com).

  • Fig. 4 Regulation of 5-hydroxytryptamine (5-HT) release by nociceptin/orphanin FQ (N/OFQ) in the dorsal raphe nucleus (DR) and frontal cortex (FC). N/OFQ and nociceptin opioid peptide (NOP) receptor are found in the DR and its terminals in the frontal cortex, medial and lateral septum, hippocampus, striatum, and nucleus accumbens. Although it is not clear whether N/OFQ is present in 5-HT neurons in the DR, NOP receptor in the DR is supposed to be located on 5-HT neurons because the injection of 5,7-dihydroxytryptamine, neurotoxin which preferentially destroys 5-HT neurons, into rat DR significantly reduces [3H]nociceptin binding and [3H]citalopram binding. N/OFQ seems to exert biphasic effect on 5-HT neurons in the DR: 5-HT neurons in the DR is tonically inhibited by nearby GABAergic interneurons. Low doses of N/OFQ are found to stimulate 5-HT neurons, and it is likely that low doses of N/OFQ hyperpolarize GABA neurons, thereby disinhibiting 5-HT neurons in the DR. On the contrary, high doses of N/OFQ inhibits 5-HT neurons by activating inwardly rectifying K+ channels through NOP receptor. This effect appears to predominate over GABAergic disinhibition effects on 5-HT neurons in the DR. This scheme is modified using Motifolio Scientific Illustration Toolkits for Presentations and Publications (www.motifolio.com).


Reference

1. Lane RM. Antidepressant drug development: Focus on triple monoamine reuptake inhibition. J Psychopharmacol. 2015; 29:526–544.
Article
2. Skolnick P, Krieter P, Tizzano J, Basile A, Popik P, Czobor P, Lippa A. Preclinical and clinical pharmacology of DOV 216,303, a “triple” reuptake inhibitor. CNS Drug Rev. 2006; 12:123–134.
3. Chen Z, Yang J, Tobak A. Designing new treatments for depression and anxiety. IDrugs. 2008; 11:189–197.
4. Witkin JM, Rorick-Kehn LM, Benvenga MJ, Adams BL, Gleason SD, Knitowski KM, Li X, Chaney S, Falcone JF, Smith JW, Foss J, Lloyd K, Catlow JT, McKinzie DL, Svensson KA, Barth VN, Toledo MA, Diaz N, Lafuente C, Jiménez A, et al. Preclinical findings predicting efficacy and side-effect profile of LY2940094, an antagonist of nociceptin receptors. Pharmacol Res Perspect. 2016; 4:e00275.
Article
5. 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. A selective nociceptin receptor antagonist to treat depression: evidence from preclinical and clinical studies. Neuropsychopharmacology. 2016; 41:1803–1812.
Article
6. Witkin JM, Wallace TL, Martin WJ. Therapeutic approaches for NOP receptor antagonists in neurobehavioral disorders: clinical studies in major depressive disorder and alcohol use disorder with BTRX-246040 (LY2940094). Handb Exp Pharmacol. 2019; 254:399–415.
Article
7. Mollereau C, Parmentier M, Mailleux P, Butour JL, Moisand C, Chalon P, Caput D, Vassart G, Meunier JC. ORL1, a novel member of the opioid receptor family. Cloning, functional expression and localization. FEBS Lett. 1994; 341:33–38.
8. Bunzow JR, Saez C, Mortrud M, Bouvier C, Williams JT, Low M, Grandy DK. 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. 1994; 347:284–288.
9. Chen Y, Fan Y, Liu J, Mestek A, Tian M, Kozak CA, Yu L. Molecular cloning, tissue distribution and chromosomal localization of a novel member of the opioid receptor gene family. FEBS Lett. 1994; 347:279–283.
Article
10. Osinski MA, Pampusch MS, Murtaugh MP, Brown DR. Cloning, expression and functional role of a nociceptin/orphanin FQ receptor in the porcine gastrointestinal tract. Eur J Pharmacol. 1999; 365:281–289.
Article
11. Meunier JC, Mollereau C, Toll L, Suaudeau C, Moisand C, Alvinerie P, Butour LC, Guillemot JC, Ferrara P, Monsarrat B, Mazarguil H, Vassart G, Parmentier M, Costentin J. Isolation and structure of the endogenous agonist of opioid receptor-like ORL1 receptor. Nature. 1995; 377:532–535.
Article
12. Reinscheid RK, Nothacker HP, Bourson A, Ardati A, Henningsen RA, Bunzow JR, Grandy DK, Langen H, Monsma FJ Jr, Civelli O. Orphanin FQ: a neuropeptide that activates an opioidlike G protein-coupled receptor. Science. 1995; 270:792–794.
Article
13. Henderson G, McKnight AT. The orphan opioid receptor and its endogenous ligand--nociceptin/orphanin FQ. Trends Pharmacol Sci. 1997; 18:293–300.
14. Gintzler AR, Adapa ID, Toll L, Medina VM, Wang L. Modulation of enkephalin release by nociceptin (orphanin FQ). Eur J Pharmacol. 1997; 325:29–34.
Article
15. Neal CR Jr, Mansour A, Reinscheid R, Nothacker HP, Civelli O, Watson SJ Jr. Localization of orphanin FQ (nociceptin) peptide and messenger RNA in the central nervous system of the rat. J Comp Neurol. 1999; 406:503–547.
Article
16. Neal CR Jr, Mansour A, Reinscheid R, Nothacker HP, Civelli O, Akil H, Watson SJ Jr. Opioid receptor-like (ORL1) receptor distribution in the rat central nervous system: comparison of ORL1 receptor mRNA expression with 125I-[14Tyr]-orphanin FQ binding. J Comp Neurol. 1999; 412:563–605.
17. Mansour A, Fox CA, Akil H, Watson SJ. Opioid-receptor mRNA expression in the rat CNS: anatomical and functional implications. Trends Neurosci. 1995; 18:22–29.
Article
18. New DC, Wong YH. The ORL1 receptor: molecular pharmacology and signalling mechanisms. Neurosignals. 2002; 11:197–212.
19. Donica CL, Awwad HO, Thakker DR, Standifer KM. Cellular mechanisms of nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor regulation and heterologous regulation by N/OFQ. Mol Pharmacol. 2013; 83:907–918.
Article
20. Zhang Z, Xin SM, Wu GX, Zhang WB, Ma L, Pei G. Endogenous delta-opioid and ORL1 receptors couple to phosphorylation and activation of p38 MAPK in NG108-15 cells and this is regulated by protein kinase A and protein kinase C. J Neurochem. 1999; 73:1502–1509.
21. Knoflach F, Reinscheid RK, Civelli O, Kemp JA. Modulation of voltage-gated calcium channels by orphanin FQ in freshly dissociated hippocampal neurons. J Neurosci. 1996; 16:6657–6664.
Article
22. Tallent MK, Madamba SG, Siggins GR. Nociceptin reduces epileptiform events in CA3 hippocampus via presynaptic and postsynaptic mechanisms. J Neurosci. 2001; 21:6940–6948.
Article
23. Levitan IB, Kaczmarek LK. The neuron: cell and molecular biology. 3rd ed. New York: Oxford University Press;2001.
24. Chin JH, Harris K, MacTavish D, Jhamandas JH. Nociceptin/orphanin FQ modulation of ionic conductances in rat basal forebrain neurons. J Pharmacol Exp Ther. 2002; 303:188–195.
Article
25. Qu L, Li Y, Tian H, Wang Z, Cui L, Jin H, Wang W, Yang L. Effects of PKC on inhibition of delayed rectifier potassium currents by N/OFQ. Biochem Biophys Res Commun. 2007; 356:582–586.
Article
26. Yao WD, Wu CF. Distinct roles of CaMKII and PKA in regulation of firing patterns and K+ currents in Drosophila neurons. J Neurophysiol. 2001; 85:1384–1394.
27. Vaughan CW, Christie MJ. Increase by the ORL1 receptor (opioid receptor-like1) ligand, nociceptin, of inwardly rectifying K conductance in dorsal raphe nucleus neurones. Br J Pharmacol. 1996; 117:1609–1611.
Article
28. Connor M, Vaughan CW, Chieng B, Christie MJ. Nociceptin receptor coupling to a potassium conductance in rat locus coeruleus neurones in vitro. Br J Pharmacol. 1996; 119:1614–1618.
29. Emmerson PJ, Miller RJ. Pre- and postsynaptic actions of opioid and orphan opioid agonists in the rat arcuate nucleus and ventromedial hypothalamus in vitro. J Physiol. 1999; 517(Pt 2):431–445.
30. Wagner EJ, Rønnekleiv OK, Grandy DK, Kelly MJ. The peptide orphanin FQ inhibits beta-endorphin neurons and neurosecretory cells in the hypothalamic arcuate nucleus by activating an inwardly-rectifying K+ conductance. Neuroendocrinology. 1998; 67:73–82.
31. Slugg RM, Rønnekleiv OK, Grandy DK, Kelly MJ. Activation of an inwardly rectifying K+ conductance by orphanin-FQ/nociceptin in vasopressin-containing neurons. Neuroendocrinology. 1999; 69:385–396.
Article
32. Chiou LC. [Phe1psi(CH2-NH)Gly2]nociceptin-(1–13)-NH2 activation of an inward rectifier as a partial agonist of ORL1 receptors in rat periaqueductal gray. Br J Pharmacol. 1999; 128:103–107.
33. Ikeda K, Kobayashi K, Kobayashi T, Ichikawa T, Kumanishi T, Kishida H, Yano R, Manabe T. Functional coupling of the nociceptin/orphanin FQ receptor with the G-protein-activated K+ (GIRK) channel. Brain Res Mol Brain Res. 1997; 45:117–126.
34. Meis S, Pape HC. Postsynaptic mechanisms underlying responsiveness of amygdaloid neurons to nociceptin/orphanin FQ. J Neurosci. 1998; 18:8133–8144.
Article
35. Farhang B, Pietruszewski L, Lutfy K, Wagner EJ. The role of the NOP receptor in regulating food intake, meal pattern, and the excitability of proopiomelanocortin neurons. Neuropharmacology. 2010; 59:190–200.
Article
36. Chee MJ, Price CJ, Statnick MA, Colmers WF. Nociceptin/orphanin FQ suppresses the excitability of neurons in the ventromedial nucleus of the hypothalamus. J Physiol. 2011; 589(Pt 13):3103–3114.
Article
37. Borgquist A, Kachani M, Tavitian N, Sinchak K, Wagner EJ. Estradiol negatively modulates the pleiotropic actions of orphanin FQ/nociceptin at proopiomelanocortin synapses. Neuroendocrinology. 2013; 98:60–72.
Article
38. Hernandez J, Fabelo C, Perez L, Moore C, Chang R, Wagner EJ. Nociceptin/orphanin FQ modulates energy homeostasis through inhibition of neurotransmission at VMN SF-1/ARC POMC synapses in a sex- and diet-dependent manner. Biol Sex Differ. 2019; 10:9.
Article
39. Xie X, Wisor JP, Hara J, Crowder TL, LeWinter R, Khroyan TV, Yamanaka A, Diano S, Horvath TL, Sakurai T, Toll L, Kilduff TS. Hypocretin/orexin and nociceptin/orphanin FQ coordinately regulate analgesia in a mouse model of stress-induced analgesia. J Clin Invest. 2008; 118:2471–2481.
Article
40. Meis S, Munsch T, Pape HC. Antioscillatory effects of nociceptin/orphanin FQ in synaptic networks of the rat thalamus. J Neurosci. 2002; 22:718–727.
Article
41. Connor M, Yeo A, Henderson G. The effect of nociceptin on Ca2+ channel current and intracellular Ca2+ in the SH-SY5Y human neuroblastoma cell line. Br J Pharmacol. 1996; 118:205–207.
42. Vaughan CW, Connor M, Jennings EA, Marinelli S, Allen RG, Christie MJ. Actions of nociceptin/orphanin FQ and other prepronociceptin products on rat rostral ventromedial medulla neurons in vitro. J Physiol. 2001; 534(Pt 3):849–859.
43. Zamponi GW, Snutch TP. Decay of prepulse facilitation of N type calcium channels during G protein inhibition is consistent with binding of a single Gbeta subunit. Proc Natl Acad Sci U S A. 1998; 95:4035–4039.
44. Pu L, Bao GB, Ma L, Pei G. Acute desensitization of nociceptin/orphanin FQ inhibition of voltage-gated calcium channels in freshly dissociated hippocampal neurons. Eur J Neurosci. 1999; 11:3610–3616.
Article
45. Connor M, Christie MJ. Modulation of Ca2+ channel currents of acutely dissociated rat periaqueductal grey neurons. J Physiol. 1998; 509(Pt 1):47–58.
46. Borgland SL, Connor M, Christie MJ. Nociceptin inhibits calcium channel currents in a subpopulation of small nociceptive trigeminal ganglion neurons in mouse. J Physiol. 2001; 536(Pt 1):35–47.
Article
47. Altier C, Khosravani H, Evans RM, Hameed S, Peloquin JB, Vartian BA, Chen L, Beedle AM, Ferguson SS, Mezghrani A, Dubel SJ, Bourinet E, McRory JE, Zamponi GW. ORL1 receptor-mediated internalization of N-type calcium channels. Nat Neurosci. 2006; 9:31–40.
Article
48. Murali SS, Napier IA, Rycroft BK, Christie MJ. Opioid-related (ORL1) receptors are enriched in a subpopulation of sensory neurons and prolonged activation produces no functional loss of surface N-type calcium channels. J Physiol. 2012; 590:1655–1667.
Article
49. Spampinato S, Di Toro R, Qasem AR. Nociceptin-induced internalization of the ORL1 receptor in human neuroblastoma cells. Neuroreport. 2001; 12:3159–3163.
Article
50. Gavioli EC, Vaughan CW, Marzola G, Guerrini R, Mitchell VA, Zucchini S, De Lima TC, Rae GA, Salvadori S, Regoli D, Calo' G. Antidepressant-like effects of the nociceptin/orphanin FQ receptor antagonist UFP-101: new evidence from rats and mice. Naunyn Schmiedebergs Arch Pharmacol. 2004; 369:547–553.
Article
51. Kawahara Y, Hesselink MB, van Scharrenburg G, Westerink BH. Tonic inhibition by orphanin FQ/nociceptin of noradrenaline neurotransmission in the amygdala. Eur J Pharmacol. 2004; 485:197–200.
Article
52. Yoshitake S, Ijiri S, Kehr J, Yoshitake T. Concurrent modulation of extracellular levels of noradrenaline and cAMP during stress and by anxiogenic- or anxiolytic-like neuropeptides in the prefrontal cortex of awake rats. Neurochem Int. 2013; 62:314–323.
Article
53. Okawa H, Kudo M, Kudo T, Guerrini R, Lambert DG, Kushikata T, Yoshida H, Matsuki A. Effects of nociceptinNH2 and [Nphe1] nociceptin(1–13)NH2 on rat brain noradrenaline release in vivo and in vitro. Neurosci Lett. 2001; 303:173–176.
54. Siniscalchi A, Rodi D, Morari M, Marti M, Cavallini S, Marino S, Beani L, Bianchi C. Direct and indirect inhibition by nociceptin/orphanin FQ on noradrenaline release from rodent cerebral cortex in vitro. Br J Pharmacol. 2002; 136:1178–1184.
55. Rominger A1, Förster S, Zentner J, Dooley DJ, McKnight AT, Feuerstein TJ, Jackisch R, Vlaskovska M. Comparison of the ORL1 receptor-mediated inhibition of noradrenaline release in human and rat neocortical slices. Br J Pharmacol. 2002; 135:800–806.
Article
56. Marti M, Stocchi S, Paganini F, Mela F, De Risi C, Calo' G, Guerrini R, Barnes TA, Lambert DG, Beani L, Bianchi C, Morari M. Pharmacological profiles of presynaptic nociceptin/orphanin FQ receptors modulating 5-hydroxytryptamine and noradrenaline release in the rat neocortex. Br J Pharmacol. 2003; 138:91–98.
Article
57. Schlicker E, Morari M. Nociceptin/orphanin FQ and neurotransmitter release in the central nervous system. Peptides. 2000; 21:1023–1029.
Article
58. Schlicker E, Werthwein S, Kathmann M, Bauer U. Nociceptin inhibits noradrenaline release in the mouse brain cortex via presynaptic ORL1 receptors. Naunyn Schmiedebergs Arch Pharmacol. 1998; 358:418–422.
Article
59. Florin S, Leroux-Nicollet I, Meunier JC, Costentin J. Autoradiographic localization of [3H]nociceptin binding sites from telencephalic to mesencephalic regions of the mouse brain. Neurosci Lett. 1997; 230:33–36.
60. Norton CS, Neal CR, Kumar S, Akil H, Watson SJ. Nociceptin/orphanin FQ and opioid receptor-like receptor mRNA expression in dopamine systems. J Comp Neurol. 2002; 444:358–368.
Article
61. Maidment NT, Chen Y, Tan AM, Murphy NP, Leslie FM. Rat ventral midbrain dopamine neurons express the orphanin FQ/nociceptin receptor ORL-1. Neuroreport. 2002; 13:1137–1140.
Article
62. Olianas MC, Dedoni S, Boi M, Onali P. Activation of nociceptin/orphanin FQ-NOP receptor system inhibits tyrosine hydroxylase phosphorylation, dopamine synthesis, and dopamine D1 receptor signaling in rat nucleus accumbens and dorsal striatum. J Neurochem. 2008; 107:544–556.
63. Lutfy K, Do T, Maidment NT. Orphanin FQ/nociceptin attenuates motor stimulation and changes in nucleus accumbens extracellular dopamine induced by cocaine in rats. Psychopharmacology (Berl). 2001; 154:1–7.
Article
64. Murphy NP, Maidment NT. Orphanin FQ/nociceptin modulation of mesolimbic dopamine transmission determined by microdialysis. J Neurochem. 1999; 73:179–186.
Article
65. Koizumi M, Sakoori K, Midorikawa N, Murphy NP. The NOP (ORL1) receptor antagonist Compound B stimulates mesolimbic dopamine release and is rewarding in mice by a non-NOP-receptor-mediated mechanism. Br J Pharmacol. 2004; 143:53–62.
Article
66. Vazquez-DeRose J, Stauber G, Khroyan TV, Xie XS, Zaveri NT, Toll L. Retrodialysis of N/OFQ into the nucleus accumbens shell blocks cocaine-induced increases in extracellular dopamine and locomotor activity. Eur J Pharmacol. 2013; 699:200–206.
Article
67. Murphy NP, Tan AM, Lam HA, Maidment NT. Nociceptin/orphanin FQ modulation of rat midbrain dopamine neurons in primary culture. Neuroscience. 2004; 127:929–940.
Article
68. Zheng F, Grandy DK, Johnson SW. Actions of orphanin FQ/nociceptin on rat ventral tegmental area neurons in vitro. Br J Pharmacol. 2002; 136:1065–1071.
69. Sesack SR, Pickel VM. Ultrastructural relationships between terminals immunoreactive for enkephalin, GABA, or both transmitters in the rat ventral tegmental area. Brain Res. 1995; 672:261–275.
Article
70. Kalivas PW, Churchill L, Klitenick MA. GABA and enkephalin projection from the nucleus accumbens and ventral pallidum to the ventral tegmental area. Neuroscience. 1993; 57:1047–1060.
Article
71. Sesack SR, Pickel VM. Dual ultrastructural localization of enkephalin and tyrosine hydroxylase immunoreactivity in the rat ventral tegmental area: multiple substrates for opiate-dopamine interactions. J Neurosci. 1992; 12:1335–1350.
Article
72. Starr MS. Multiple opiate receptors may be involved in suppressing gamma-aminobutyrate release in substantia nigra. Life Sci. 1985; 37:2249–2255.
73. Tanaka E, North RA. Opioid actions on rat anterior cingulate cortex neurons in vitro. J Neurosci. 1994; 14(3 Pt 1):1106–1113.
74. Mogil JS, Grisel JE, Zhangs G, Belknap JK, Grandy DK. Functional antagonism of mu-, delta- and kappa-opioid antinociception by orphanin FQ. Neurosci Lett. 1996; 214:131–134.
75. Van Bockstaele EJ, Pickel VM. GABA-containing neurons in the ventral tegmental area project to the nucleus accumbens in rat brain. Brain Res. 1995; 682:215–221.
Article
76. Anton B, Fein J, To T, Li X, Silberstein L, Evans CJ. Immunohistochemical localization of ORL-1 in the central nervous system of the rat. J Comp Neurol. 1996; 368:229–251.
Article
77. Le Maître E, Vilpoux C, Costentin J, Leroux-Nicollet I. Opioid receptor-like 1 (NOP) receptors in the rat dorsal raphe nucleus: evidence for localization on serotoninergic neurons and functional adaptation after 5,7-dihydroxytryptamine lesion. J Neurosci Res. 2005; 81:488–496.
Article
78. Nazzaro C, Barbieri M, Varani K, Beani L, Valentino RJ, Siniscalchi A. Swim stress enhances nociceptin/orphanin FQ-induced inhibition of rat dorsal raphe nucleus activity in vivo and in vitro: role of corticotropin releasing factor. Neuropharmacology. 2010; 58:457–464.
79. Nazzaro C, Marino S, Barbieri M, Siniscalchi A. Inhibition of serotonin outflow by nociceptin/orphaninFQ in dorsal raphe nucleus slices from normal and stressed rats: role of corticotropin releasing factor. Neurochem Int. 2009; 54:378–384.
Article
80. Tao R, Ma Z, Thakkar MM, McCarley RW, Auerbach SB. Nociceptin/orphanin FQ decreases serotonin efflux in the rat brain but in contrast to a kappa-opioid has no antagonistic effect on muopioid-induced increases in serotonin efflux. Neuroscience. 2007; 147:106–116.
81. Le Maître E, Dourmap N, Vilpoux C, Leborgne R, Janin F, Bonnet JJ, Costentin J, Leroux-Nicollet I. Acute and subchronic treatments with selective serotonin reuptake inhibitors increase Nociceptin/Orphanin FQ (NOP) receptor density in the rat dorsal raphe nucleus; interactions between nociceptin/NOP system and serotonin. Brain Res. 2013; 1520:51–60.
Article
82. Sbrenna S, Marti M, Morari M, Calo' G, Guerrini R, Beani L, Bianchi C. Modulation of 5-hydroxytryptamine efflux from rat cortical synaptosomes by opioids and nociceptin. Br J Pharmacol. 2000; 130:425–433.
Article
83. Calo G, Rizzi A, Rizzi D, Bigoni R, Guerrini R, Marzola G, Marti M, McDonald J, Morari M, Lambert DG, Salvadori S, Regoli D. [Nphe1,Arg14,Lys15]nociceptin-NH2, a novel potent and selective antagonist of the nociceptin/orphanin FQ receptor. Br J Pharmacol. 2002; 136:303–311.
84. Werthwein S, Bauer U, Nakazi M, Kathmann M, Schlicker E. Further characterization of the ORL1 receptor-mediated inhibition of noradrenaline release in the mouse brain in vitro. Br J Pharmacol. 1999; 127:300–308.
85. Berger B, Rothmaier AK, Wedekind F, Zentner J, Feuerstein TJ, Jackisch R. Presynaptic opioid receptors on noradrenergic and serotonergic neurons in the human as compared to the rat neocortex. Br J Pharmacol. 2006; 148:795–806.
Article
86. Devine DP, Watson SJ, Akil H. Nociceptin/orphanin FQ regulates neuroendocrine function of the limbic-hypothalamic-pituitary-adrenal axis. Neuroscience. 2001; 102:541–553.
Article
87. Leggett JD, Harbuz MS, Jessop DS, Fulford AJ. The nociceptin receptor antagonist [Nphe1,Arg14,Lys15]nociceptin/orphanin FQ-NH2 blocks the stimulatory effects of nociceptin/orphanin FQ on the HPA axis in rats. Neuroscience. 2006; 141:2051–2057.
88. Nicholson JR, Akil H, Watson SJ. Orphanin FQ-induced hyperphagia is mediated by corticosterone and central glucocorticoid receptors. Neuroscience. 2002; 115:637–643.
Article
89. Le Cudennec C, Naudin B, Do Rego JC, Costentin J. Nociceptin/orphanin FQ and related peptides reduce the increase in plasma corticosterone elicited in mice by an intracerebroventricular injection. Life Sci. 2002; 72:163–171.
Article
90. Fernandez F, Misilmeri MA, Felger JC, Devine DP. Nociceptin/orphanin FQ increases anxiety-related behavior and circulating levels of corticosterone during neophobic tests of anxiety. Neuropsychopharmacology. 2004; 29:59–71.
Article
91. Green MK, Barbieri EV, Brown BD, Chen KW, Devine DP. Roles of the bed nucleus of stria terminalis and of the amygdala in N/OFQ-mediated anxiety and HPA axis activation. Neuropeptides. 2007; 41:399–410.
Article
92. Gottlieb HB, Fleming TM, Ji L, Cunningham JT. Identification of central nervous system sites involved in the water diuresis response elicited by central microinjection of nociceptin/Orphanin FQ in conscious rats via c-Fos and inducible cAMP early repressor immunocytochemistry. J Neuroendocrinol. 2007; 19:531–542.
93. Kazi JA. Nocistatin and nociceptin modulate c-Fos expression in the mice thalamus. Neurol Sci. 2012; 33:1233–1237.
Article
94. Olszewski PK, Billington CJ, Levine AS. Fos expression in feeding-related brain areas following intracerebroventricular administration of orphanin FQ in rats. Brain Res. 2000; 855:171–175.
Article
95. Leggett JD, Dawe KL, Jessop DS, Fulford AJ. Endogenous nociceptin/orphanin FQ system involvement in hypothalamicpituitary-adrenal axis responses: relevance to models of inflammation. J Neuroendocrinol. 2009; 21:888–897.
Article
96. Kawashima N, Fugate J, Kusnecov AW. Immunological challenge modulates brain orphanin FQ/nociceptin and nociceptive behavior. Brain Res. 2002; 949:71–78.
Article
97. Nativio P, Pascale E, Maffei A, Scaccianoce S, Passarelli F. Effect of stress on hippocampal nociceptin expression in the rat. Stress. 2012; 15:378–384.
Article
98. Delaney G, Dawe KL, Hogan R, Hunjan T, Roper J, Hazell G, Lolait SJ, Fulford AJ. Role of nociceptin/orphanin FQ and NOP receptors in the response to acute and repeated restraint stress in rats. J Neuroendocrinol. 2012; 24:1527–1541.
99. Green MK, Devine DP. Nociceptin/orphanin FQ and NOP receptor gene regulation after acute or repeated social defeat stress. Neuropeptides. 2009; 43:507–514.
Article
100. Martinez M, Phillips PJ, Herbert J. Adaptation in patterns of c-fos expression in the brain associated with exposure to either single or repeated social stress in male rats. Eur J Neurosci. 1998; 10:20–33.
101. Pan YX, Xu J, Pasternak GW. Structure and characterization of the gene encoding a mouse kappa3-related opioid receptor. Gene. 1996; 171:255–260.
Article
102. Xie GX, Ito E, Maruyama K, Suzuki Y, Sugano S, Sharma M, Pietruck C, Palmer PP. The promoter region of human prepronociceptin gene and its regulation by cyclic AMP and steroid hormones. Gene. 1999; 238:427–436.
Article
103. Granholm L, Roman E, Nylander I. Single housing during early adolescence causes time-, area- and peptide-specific alterations in endogenous opioids of rat brain. Br J Pharmacol. 2015; 172:606–614.
Article
104. Tolchard S, Hare AS, Nutt DJ, Clarke G. TNF alpha mimics the endocrine but not the thermoregulatory responses of bacterial lipopolysaccharide (LPS): correlation with FOS-expression in the brain. Neuropharmacology. 1996; 35:243–248.
105. Devine DP, Hoversten MT, Ueda Y, Akil H. Nociceptin/orphanin FQ content is decreased in forebrain neurones during acute stress. J Neuroendocrinol. 2003; 15:69–74.
Article
106. Ploj K, Roman E, Nylander I. Effects of maternal separation on brain nociceptin/orphanin FQ peptide levels in male Wistar rats. Pharmacol Biochem Behav. 2002; 73:123–129.
Article
107. Reiss D, Wolter-Sutter A, Krezel W, Ouagazzal AM. Effects of social crowding on emotionality and expression of hippocampal nociceptin/orphanin FQ system transcripts in mice. Behav Brain Res. 2007; 184:167–173.
Article
108. Alder J, Kallman S, Palmieri A, Khadim F, Ayer JJ, Kumar S, Tsung K, Grinberg I, Thakker-Varia S. Neuropeptide orphanin FQ inhibits dendritic morphogenesis through activation of RhoA. Dev Neurobiol. 2013; 73:769–784.
Article
109. Ring RH, Alder J, Fennell M, Kouranova E, Black IB, Thakker-Varia S. Transcriptional profiling of brain-derived-neurotrophic factor-induced neuronal plasticity: a novel role for nociceptin in hippocampal neurite outgrowth. J Neurobiol. 2006; 66:361–377.
Article
110. Gavioli EC, Marzola G, Guerrini R, Bertorelli R, Zucchini S, De Lima TC, Rae GA, Salvadori S, Regoli D, Calo G. 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. 2003; 17:1987–1990.
Article
111. Redrobe JP, Calo' G, Regoli D, Quirion R. Nociceptin receptor antagonists display antidepressant-like properties in the mouse forced swimming test. Naunyn Schmiedebergs Arch Pharmacol. 2002; 365:164–167.
Article
112. Holanda VAD, Santos WB, Asth L, Guerrini R, Calo' G, Ruzza C, Gavioli EC. NOP agonists prevent the antidepressant-like effects of nortriptyline and fluoxetine but not R-ketamine. Psychopharmacology (Berl). 2018; 235:3093–3102.
Article
113. Rizzi A, Gavioli EC, Marzola G, Spagnolo B, Zucchini S, Ciccocioppo R, Trapella C, Regoli D, Calò G. 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. 2007; 321:968–974.
114. Holanda VA, Medeiros IU, Asth L, Guerrini R, Calo' G, Gavioli EC. Antidepressant activity of nociceptin/orphanin FQ receptor antagonists in the mouse learned helplessness. Psychopharmacology (Berl). 2016; 233:2525–2532.
Article
115. Medeiros IU, Ruzza C, Asth L, Guerrini R, Romão PR, Gavioli EC, Calo G. Blockade of nociceptin/orphanin FQ receptor signaling reverses LPS-induced depressive-like behavior in mice. Peptides. 2015; 72:95–103.
Article
116. Vitale G, Filaferro M, Micioni Di Bonaventura MV, Ruggieri V, Cifani C, Guerrini R, Simonato M, Zucchini S. Effects of [Nphe1, Arg14, Lys15] N/OFQ-NH2 (UFP-101), a potent NOP receptor antagonist, on molecular, cellular and behavioural alterations associated with chronic mild stress. J Psychopharmacol. 2017; 31:691–703.
117. Vitale G, Ruggieri V, Filaferro M, Frigeri C, Alboni S, Tascedda F, Brunello N, Guerrini R, Cifani C, Massi M. Chronic treatment with the selective NOP receptor antagonist [Nphe1,Arg14,Lys15]N/OFQ-NH2 (UFP-101) reverses the behavioural and biochemical effects of unpredictable chronic mild stress in rats. Psychopharmacology (Berl). 2009; 207:173–189.
118. Gu H, Hu D, Hong XR, Mao J, Cui Y, Hui N, Sha JY. Changes and significance of orphanin and serotonin in patients with postpartum depression. Zhonghua Fu Chan Ke Za Zhi. 2003; 38:727–728.
119. Wang LN, Liu LF, Zhang JX, Zhao GF. Plasma levels of nociceptin/orphanin FQ in patients with bipolar disorders and health adults. Zhonghua Yi Xue Za Zhi. 2009; 89:916–918.
120. Aziz AM, Brothers S, Sartor G, Holm L, Heilig M, Wahlestedt C, Thorsell A. The nociceptin/orphanin FQ receptor agonist SR-8993 as a candidate therapeutic for alcohol use disorders: validation in rat models. Psychopharmacology (Berl). 2016; 233:3553–3563.
Article
121. aan het Rot M, Mathew SJ, Charney DS. Neurobiological mechanisms in major depressive disorder. CMAJ. 2009; 180:305–313.
Article
122. Nestler EJ, Barrot M, DiLeone RJ, Eisch AJ, Gold SJ, Monteggia LM. Neurobiology of depression. Neuron. 2002; 34:13–25.
Article
123. Detke MJ, Rickels M, Lucki I. Active behaviors in the rat forced swimming test differentially produced by serotonergic and noradrenergic antidepressants. Psychopharmacology (Berl). 1995; 121:66–72.
Article
124. Hughes ZA, Stanford SC. A partial noradrenergic lesion induced by DSP-4 increases extracellular noradrenaline concentration in rat frontal cortex: a microdialysis study in vivo. Psychopharmacology (Berl). 1998; 136:299–303.
125. Cryan JF, Page ME, Lucki I. Noradrenergic lesions differentially alter the antidepressant-like effects of reboxetine in a modified forced swim test. Eur J Pharmacol. 2002; 436:197–205.
Article
126. Kirby LG, Allen AR, Lucki I. Regional differences in the effects of forced swimming on extracellular levels of 5-hydroxytryptamine and 5-hydroxyindoleacetic acid. Brain Res. 1995; 682:189–196.
Article
127. Mangiavacchi S, Masi F, Scheggi S, Leggio B, De Montis MG, Gambarana C. Long-term behavioral and neurochemical effects of chronic stress exposure in rats. J Neurochem. 2001; 79:1113–1121.
Article
128. Ressler KJ, Nemeroff CB. Role of serotonergic and noradrenergic systems in the pathophysiology of depression and anxiety disorders. Depress Anxiety. 2000; 12 Suppl 1:2–19.
Article
129. Rosa-Neto P, Diksic M, Okazawa H, Leyton M, Ghadirian N, Mzengeza S, Nakai A, Debonnel G, Blier P, Benkelfat C. Measurement of brain regional alpha-[11C]methyl-L-tryptophan trapping as a measure of serotonin synthesis in medication-free patients with major depression. Arch Gen Psychiatry. 2004; 61:556–563.
130. Arnone D, McIntosh AM, Ebmeier KP, Munafò MR, Anderson IM. Magnetic resonance imaging studies in unipolar depression: systematic review and meta-regression analyses. Eur Neuropsychopharmacol. 2012; 22:1–16.
Article
131. Cole J, Costafreda SG, McGuffin P, Fu CH. Hippocampal atrophy in first episode depression: a meta-analysis of magnetic resonance imaging studies. J Affect Disord. 2011; 134:483–487.
Article
132. Zhang FF, Peng W, Sweeney JA, Jia ZY, Gong QY. Brain structure alterations in depression: Psychoradiological evidence. CNS Neurosci Ther. 2018; 24:994–1003.
Article
133. Kang HJ, Voleti B, Hajszan T, Rajkowska G, Stockmeier CA, Licznerski P, et al. Decreased expression of synapse-related genes and loss of synapses in major depressive disorder. Nat Med. 2012; 18:1413–1417.
Article
134. Rajkowska G, Miguel-Hidalgo JJ, Wei J, Dilley G, Pittman SD, Meltzer HY, Overholser JC, Roth BL, Stockmeier CA. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol Psychiatry. 1999; 45:1085–1098.
135. Stockmeier CA, Mahajan GJ, Konick LC, Overholser JC, Jurjus GJ, Meltzer HY, et al. Cellular changes in the postmortem hippocampus in major depression. Biol Psychiatry. 2004; 56:640–650.
Article
136. Sheline YI, Gado MH, Kraemer HC. Untreated depression and hippocampal volume loss. Am J Psychiatry. 2003; 160:1516–1518.
Article
137. Duman RS, Aghajanian GK. Synaptic dysfunction in depression: potential therapeutic targets. Science. 2012; 338:68–72.
Article
138. McEwen BS, Nasca C, Gray JD. Stress effects on neuronal structure: hippocampus, amygdala, and prefrontal cortex. Neuropsychopharmacology. 2016; 41:3–23.
Article
139. Castrén E, Kojima M. Brain-derived neurotrophic factor in mood disorders and antidepressant treatments. Neurobiol Dis. 2017; 97(Pt B):119–126.
Article
140. Levy MJF, Boulle F, Steinbusch HW, van den Hove DLA, Kenis G, Lanfumey L. Neurotrophic factors and neuroplasticity pathways in the pathophysiology and treatment of depression. Psychopharmacology (Berl). 2018; 235:2195–2220.
Article
141. Dunham JS, Deakin JFW, Miyajima F, Payton A, Toro CT. Expression of hippocampal brain-derived neurotrophic factor and its receptors in Stanley consortium brains. J Psychiatr Res. 2009; 43:1175–1184.
Article
142. Pandey GN, Ren X, Rizavi HS, Conley RR, Roberts RC, Dwivedi Y. Brain-derived neurotrophic factor and tyrosine kinase B receptor signalling in post-mortem brain of teenage suicide victims. Int J Neuropsychopharmacol. 2008; 11:1047–1061.
Article
143. Gaughran F, Payne J, Sedgwick PM, Cotter D, Berry M. Hippocampal FGF-2 and FGFR1 mRNA expression in major depression, schizophrenia and bipolar disorder. Brain Res Bull. 2006; 70:221–227.
Article
144. Chen B, Dowlatshahi D, MacQueen GM, Wang JF, Young LT. Increased hippocampal BDNF immunoreactivity in subjects treated with antidepressant medication. Biol Psychiatry. 2001; 50:260–265.
Article
145. Evans SJ, Choudary PV, Neal CR, Li JZ, Vawter MP, Tomita H, Lopez JF, Thompson RC, Meng F, Stead JD, Walsh DM, Myers RM, Bunney WE, Watson SJ, Jones EG, Akil H. Dysregulation of the fibroblast growth factor system in major depression. Proc Natl Acad Sci U S A. 2004; 101:15506–15511.
Article
146. Deng Z, Deng S, Zhang MR, Tang MM. Fibroblast growth factors in depression. Front Pharmacol. 2019; 10:60.
Article
147. Elsayed M, Banasr M, Duric V, Fournier NM, Licznerski P, Duman RS. Antidepressant effects of fibroblast growth factor-2 in behavioral and cellular models of depression. Biol Psychiatry. 2012; 72:258–265.
Article
148. Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman RS. Brain-derived neurotrophic factor produces antidepressant effects in behavioral models of depression. J Neurosci. 2002; 22:3251–3261.
Article
149. Turner CA, Gula EL, Taylor LP, Watson SJ, Akil H. Antidepressant-like effects of intracerebroventricular FGF2 in rats. Brain Res. 2008; 1224:63–68.
Article
150. Bachis A, Mallei A, Cruz MI, Wellstein A, Mocchetti I. Chronic antidepressant treatments increase basic fibroblast growth factor and fibroblast growth factor-binding protein in neurons. Neuropharmacology. 2008; 55:1114–1120.
Article
151. Mallei A, Shi B, Mocchetti I. Antidepressant treatments induce the expression of basic fibroblast growth factor in cortical and hippocampal neurons. Mol Pharmacol. 2002; 61:1017–1024.
Article
152. Nibuya M, Morinobu S, Duman RS. Regulation of BDNF and trkB mRNA in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci. 1995; 15:7539–7547.
Article
153. Zhang Y, Gu F, Chen J, Dong W. Chronic antidepressant administration alleviates frontal and hippocampal BDNF deficits in CUMS rat. Brain Res. 2010; 1366:141–148.
Article
154. Monteggia LM, Barrot M, Powell CM, Berton O, Galanis V, Gemelli T, Meuth S, Nagy A, Greene RW, Nestler EJ. Essential role of brain-derived neurotrophic factor in adult hippocampal function. Proc Natl Acad Sci U S A. 2004; 101:10827–10832.
Article
155. Saarelainen T, Hendolin P, Lucas G, Koponen E, Sairanen M, Mac-Donald E, Agerman K, Haapasalo A, Nawa H, Aloyz R, Ernfors P, Castrén E. Activation of the TrkB neurotrophin receptor is induced by antidepressant drugs and is required for antidepressant-induced behavioral effects. J Neurosci. 2003; 23:349–357.
Article
156. Simard S, Shail P, MacGregor J, El Sayed M, Duman RS, Vaccarino FM, Salmaso N. Fibroblast growth factor 2 is necessary for the antidepressant effects of fluoxetine. PLoS One. 2018; 13:e0204980.
Article
157. Pariante CM. Risk factors for development of depression and psychosis. glucocorticoid receptors and pituitary implications for treatment with antidepressant and glucocorticoids. Ann N Y Acad Sci. 2009; 1179:144–152.
158. Kvarta MD, Bradbrook KE, Dantrassy HM, Bailey AM, Thompson SM. Corticosterone mediates the synaptic and behavioral effects of chronic stress at rat hippocampal temporoammonic synapses. J Neurophysiol. 2015; 114:1713–1724.
Article
159. Gourley SL, Taylor JR. Recapitulation and reversal of a persistent depression-like syndrome in rodents. Curr Protoc Neurosci. 2009; Chapter 9:Unit 9.32.
Article
160. Gourley SL, Wu FJ, Kiraly DD, Ploski JE, Kedves AT, Duman RS, Taylor JR. Regionally specific regulation of ERK MAP kinase in a model of antidepressant-sensitive chronic depression. Biol Psychiatry. 2008; 63:353–359.
Article
161. Johnson SA, Fournier NM, Kalynchuk LE. Effect of different doses of corticosterone on depression-like behavior and HPA axis responses to a novel stressor. Behav Brain Res. 2006; 168:280–288.
Article
162. Wulsin AC, Herman JP, Solomon MB. Mifepristone decreases depression-like behavior and modulates neuroendocrine and central hypothalamic-pituitary-adrenocortical axis responsiveness to stress. Psychoneuroendocrinology. 2010; 35:1100–1112.
Article
163. Solomon MB, Wulsin AC, Rice T, Wick D, Myers B, McKlveen J, Flak JN, Ulrich-Lai Y, Herman JP. The selective glucocorticoid receptor antagonist CORT 108297 decreases neuroendocrine stress responses and immobility in the forced swim test. Horm Behav. 2014; 65:363–371.
Article
164. Wu LM, Han H, Wang QN, Hou HL, Tong H, Yan XB, Zhou JN. Mifepristone repairs region-dependent alteration of synapsin I in hippocampus in rat model of depression. Neuropsychopharmacology. 2007; 32:2500–2510.
Article
165. Block TS, Kushner H, Kalin N, Nelson C, Belanoff J, Schatzberg A. Combined analysis of mifepristone for psychotic depression: plasma levels associated with clinical response. Biol Psychiatry. 2018; 84:46–54.
Article
166. Lucassen PJ, Oomen CA, Naninck EF, Fitzsimons CP, van Dam AM, Czeh B, Korosi A. Regulation of adult neurogenesis and plasticity by (early) stress, glucocorticoids, and inflammation. Cold Spring Harb Perspect Biol. 2015; 7:a021303.
Article
167. Oomen CA, Mayer JL, de Kloet ER, Joëls M, Lucassen PJ. Brief treatment with the glucocorticoid receptor antagonist mifepristone normalizes the reduction in neurogenesis after chronic stress. Eur J Neurosci. 2007; 26:3395–3401.
Article
168. Mayer JL, Klumpers L, Maslam S, de Kloet ER, Joëls M, Lucassen PJ. Brief treatment with the glucocorticoid receptor antagonist mifepristone normalises the corticosterone-induced reduction of adult hippocampal neurogenesis. J Neuroendocrinol. 2006; 18:629–631.
Article
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr