493
Views
1
CrossRef citations to date
0
Altmetric
Review Articles

Imaging corticotropin-releasing-factor and nociceptin in addiction and PTSD models

, &
Pages 567-579 | Received 26 Oct 2017, Accepted 08 Nov 2017, Published online: 12 Dec 2017

References

  • Akana, S. F., Dallman, M. F., Bradbury, M. J., Scribner, K. A., Strack, A. M., & Walker, C. D. (1992). Feedback and facilitation in the adrenocortical system: unmasking facilitation by partial inhibition of the glucocorticoid response to prior stress. Endocrinology, 131, 57–68. doi:10.1210/endo.131.1.1319329
  • Andero, R., Brothers, S. P., Jovanovic, T., Chen, Y. T., Salah-Uddin, H., Cameron, M., … Ressler, K. J. (2013). Amygdala-dependent fear is regulated by Oprl1 in mice and humans with PTSD. Science Translational Medicine, 5, 188ra173. doi:10.1126/scitranslmed.3005656
  • Baur, V., Hanggi, J., Langer, N., & Jancke, L. (2013). Resting-state functional and structural connectivity within an insula-amygdala route specifically index state and trait anxiety. Biological Psychiatry, 73, 85–92. doi:10.1016/j.biopsych.2012.06.003
  • Berenz, E. C., & Coffey, S. F. (2012). Treatment of co-occurring posttraumatic stress disorder and substance use disorders. Current Psychiatry Reports, 14, 469–477. doi:10.1007/s11920-012-0300-0
  • Berthele, A., Platzer, S., Dworzak, D., Schadrack, J., Mahal, B., Buttner, A., … Tolle, T. R. (2003). [3H]-nociceptin ligand-binding and nociceptin opioid receptor mrna expression in the human brain. Neuroscience, 121, 629–640.
  • Bornhovd, K., Quante, M., Glauche, V., Bromm, B., Weiller, C., & Buchel, C. (2002). Painful stimuli evoke different stimulus-response functions in the amygdala, prefrontal, insula and somatosensory cortex: a single-trial fMRI study. Brain, 125, 1326–1336. doi:10.1093/brain/awf137
  • Bridge, K. E., Wainwright, A., Reilly, K., & Oliver, K. R. (2003). Autoradiographic localization of (125)i[Tyr(14)] nociceptin/orphanin FQ binding sites in macaque primate CNS. Neuroscience, 118, 513–523. doi:10.1016/S0306-4522(02)00927-2
  • Ciccocioppo, R., de Guglielmo, G., Hansson, A. C., Ubaldi, M., Kallupi, M., Cruz, M. T., … Roberto, M. (2014). Restraint stress alters nociceptin/orphanin FQ and CRF systems in the rat central amygdala: significance for anxiety-like behaviors. Journal of Neuroscience, 34, 363–372. doi:10.1523/JNEUROSCI.2400-13.2014
  • Ciccocioppo, R., Economidou, D., Fedeli, A., Angeletti, S., Weiss, F., Heilig, M., & Massi, M. (2004). Attenuation of ethanol self-administration and of conditioned reinstatement of alcohol-seeking behaviour by the antiopioid peptide nociceptin/orphanin FQ in alcohol-preferring rats. Psychopharmacology (Berl), 172, 170–178. doi:10.1007/s00213-003-1645-1
  • Ciccocioppo, R., Fedeli, A., Economidou, D., Policani, F., Weiss, F., & Massi, M. (2003). The bed nucleus is a neuroanatomical substrate for the anorectic effect of corticotropin-releasing factor and for its reversal by nociceptin/orphanin FQ. Journal of Neuroscience, 23, 9445–9451.
  • Ciccocioppo, R., Panocka, I., Polidori, C., Regoli, D., & Massi, M. (1999). Effect of nociceptin on alcohol intake in alcohol-preferring rats. Psychopharmacology (Berl), 141, 220–224. doi:10.1007/s002130050828
  • Ciccocioppo, R., Polidori, C., Antonelli, L., Salvadori, S., Guerrini, R., & Massi, M. (2002). Pharmacological characterization of the nociceptin receptor which mediates reduction of alcohol drinking in rats. Peptides, 23, 117–125. doi:10.1016/S0196-9781(01)00587-3
  • Cook, C. J. (2004). Stress induces CRF release in the paraventricular nucleus, and both CRF and GABA release in the amygdala. Physiology & Behavior, 82, 751–762. doi:10.1016/j.physbeh.2004.06.013
  • Delaney, G., Dawe, K. L., Hogan, R., Hunjan, T., Roper, J., Hazell, G., … Fulford, A. J. (2012). Role of nociceptin/orphanin FQ and NOP receptors in the response to acute and repeated restraint stress in rats. Journal of Neuroendocrinology, 24, 1527–1541. doi:10.1111/j.1365-2826.2012.02361.x
  • Economidou, D., Fedeli, A., Fardon, R. M., Weiss, F., Massi, M., & Ciccocioppo, R. (2006). Effect of novel nociceptin/orphanin FQ-NOP receptor ligands on ethanol drinking in alcohol-preferring msP rats. Peptides, 27, 3299–3306. doi:10.1016/j.peptides.2006.09.007
  • Economidou, D., Hansson, A. C., Weiss, F., Terasmaa, A., Sommer, W. H., Cippitelli, A., … Heilig, M. (2008). Dysregulation of nociceptin/orphanin FQ activity in the amygdala is linked to excessive alcohol drinking in the rat. Biological Psychiatry, 64, 211–218. doi:10.1016/j.biopsych.2008.02.004
  • Enman, N. M., Zhang, Y., & Unterwald, E. M. (2014). Connecting the pathology of posttraumatic stress and substance use disorders: monoamines and neuropeptides. Pharmacology Biochemistry and Behavior, 117, 61–69. doi:10.1016/j.pbb.2013.12.001
  • Furay, A. R., Bruestle, A. E., & Herman, J. P. (2008). The role of the forebrain glucocorticoid receptor in acute and chronic stress. Endocrinology, 149, 5482–5490. doi:10.1210/en.2008-0642
  • Herman, J. P., & Mueller, N. K. (2006). Role of the ventral subiculum in stress integration. Behavioural Brain Research, 174, 215–224. doi:10.1016/j.bbr.2006.05.035
  • Hostetler, E. D., Sanabria-Bohorquez, S., Eng, W., Joshi, A. D., Patel, S., Gibson, R. E., … Hargreaves, R. J. (2013). Evaluation of [(1)(8)F]MK-0911, a positron emission tomography (PET) tracer for opioid receptor-like 1 (ORL1), in rhesus monkey and human. NeuroImage, 68, 1–10. doi:10.1016/j.neuroimage.2012.11.053
  • Jenck, F., Moreau, J. L., Martin, J. R., Kilpatrick, G. J., Reinscheid, R. K., Monsma, F. J., Jr., … Civelli, O. (1997). Orphanin FQ acts as an anxiolytic to attenuate behavioral responses to stress. Proceedings of the National Academy of Sciences of the United States of America, 94, 14854–14858.
  • Kenakin, T. (2011). Functional selectivity and biased receptor signaling. Journal of Pharmacology and Experimental Therapeutics, 336, 296–302. doi:10.1124/jpet.110.173948
  • Kessler, R. C., Berglund, P., Demler, O., Jin, R., Merikangas, K. R., & Walters, E. E. (2005). Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the national comorbidity survey replication. Archives of General Psychiatry, 62, 593–602. doi:10.1001/archpsyc.62.6.593
  • Kessler, R. C., Birnbaum, H., Demler, O., Falloon, I. R., Gagnon, E., Guyer, M., … Wu, E. Q. (2005). The prevalence and correlates of nonaffective psychosis in the national comorbidity survey replication (NCS-R). Biological Psychiatry, 58, 668–676. doi:10.1016/j.biopsych.2005.04.034
  • Kimura, Y., Fujita, M., Hong, J., Lohith, T. G., Gladding, R. L., Zoghbi, S. S., … Innis, R. B. (2011). Brain and whole-body imaging in rhesus monkeys of 11C-NOP-1A, a promising PET radioligand for nociceptin/orphanin FQ peptide receptors. Journal of Nuclear Medicine, 52, 1638–1645. doi:10.2967/jnumed.111.091181
  • Koob, G. F. (2004). Allostatic view of motivation: implications for psychopathology. Nebraska Symposium on Motivation, 50, 1–18.
  • Koob, G. F. (2008). A role for brain stress systems in addiction. Neuron, 59, 11–34. doi:10.1016/j.neuron.2008.06.012
  • Koob, G. F. (2009). Neurobiological substrates for the dark side of compulsivity in addiction. Neuropharmacology, 56(Suppl 1), 18–31. doi:10.1016/j.neuropharm.2008.07.043
  • Koob, G. F., & Heinrichs, S. C. (1999). A role for corticotropin releasing factor and urocortin in behavioral responses to stressors. Brain Research, 848, 141–152. doi:10.1016/S0006-8993(99)01991-5
  • Koob, G. F., & Le Moal, M. (2001). Drug addiction, dysregulation of reward, and allostasis. Neuropsychopharmacology, 24, 97–129. doi:10.1016/S0893-133X(00)00195-0
  • Koster, A., Montkowski, A., Schulz, S., Stube, E. M., Knaudt, K., Jenck, F., … Reinscheid, R. K. (1999). Targeted disruption of the orphanin FQ/nociceptin gene increases stress susceptibility and impairs stress adaptation in mice. Proceedings of the National Academy of Sciences of the United States of America, 96, 10444–10449.
  • Kuzmin, A., Bazov, I., Sheedy, D., Garrick, T., Harper, C., & Bakalkin, G. (2009). Expression of pronociceptin and its receptor is downregulated in the brain of human alcoholics. Brain Research, 1305(Suppl), S80–S85. doi:10.1016/j.brainres.2009.05.067
  • LeDoux, J. E. (2000). Emotion circuits in the brain. Annual Review of Neuroscience, 23, 155–184. doi:10.1146/annurev.neuro.23.1.155
  • Leggett, J. D., Harbuz, M. S., Jessop, D. S., & Fulford, A. J. (2006). 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, 141, 2051–2057. doi:10.1016/j.neuroscience.2006.05.036
  • Leggett, J. D., Jessop, D. S., & Fulford, A. J. (2007). The nociceptin/orphanin FQ antagonist UFP-101 differentially modulates the glucocorticoid response to restraint stress in rats during the peak and nadir phases of the hypothalamo-pituitary-adrenal axis circadian rhythm. Neuroscience, 147, 757–764. doi:10.1016/j.neuroscience.2007.04.010
  • Lodge, N. J., Li, Y. W., Chin, F. T., Dischino, D. D., Zoghbi, S. S., Deskus, J. A., … Pike, V. W. (2014). Synthesis and evaluation of candidate PET radioligands for corticotropin-releasing factor type-1 receptors. Nuclear Medicine and Biology, 41, 524–535. doi:10.1016/j.nucmedbio.2014.03.005
  • Lohith, T. G., Zoghbi, S. S., Morse, C. L., Araneta, M. D., Barth, V. N., Goebl, N. A., … Fujita, M. (2014). Retest imaging of [11C]NOP-1A binding to nociceptin/orphanin FQ peptide (NOP) receptors in the brain of healthy humans. Neuroimage, 87, 89–95. doi:10.1016/j.neuroimage.2013.10.068
  • Lohith, T. G., Zoghbi, S. S., Morse, C. L., Araneta, M. F., Barth, V. N., Goebl, N. A., … Fujita, M. (2012). Brain and whole-body imaging of nociceptin/orphanin FQ peptide receptor in humans using the PET ligand 11C-NOP-1A. Journal of Nuclear Medicine, 53, 385–392. doi:10.2967/jnumed.111.097162
  • Mallimo, E. M., & Kusnecov, A. W. (2013). The role of orphanin FQ/nociceptin in neuroplasticity: relationship to stress, anxiety and neuroinflammation. Frontiers in Cellular Neuroscience, 7, 173. doi:10.3389/fncel.2013.00173
  • Martin-Fardon, R., Ciccocioppo, R., Massi, M., & Weiss, F. (2000). Nociceptin prevents stress-induced ethanol- but not cocaine-seeking behavior in rats. Neuroreport, 11, 1939–1943. doi:10.1097/00001756-200006260-00026
  • Narendran, R., Ciccocioppo, R., Lopresti, B., Paris, J., Himes, M. L., & Mason, N. S. (2017). Nociceptin receptors in alcohol use disorders: a positron emission tomography study using [11C]NOP-1A. Biological Psychiatry, doi:10.1016/j.biopsych.2017.05.019
  • Neugebauer, V., Li, W., Bird, G. C., & Han, J. S. (2004). The amygdala and persistent pain. Neuroscientist, 10, 221–234. doi:10.1177/1073858403261077
  • Pike, V. W., Rash, K. S., Chen, Z., Pedregal, C., Statnick, M. A., Kimura, Y., … Innis, R. B. (2011). Synthesis and evaluation of radioligands for imaging brain nociceptin/orphanin FQ peptide (NOP) receptors with positron emission tomography. Journal of Medicinal Chemistry, 54, 2687–2700. doi:10.1021/jm101487v
  • Post, A., Smart, T. S., Jackson, K., Mann, J., Mohs, R., Rorick-Kehn, L., … Wong, C. J. (2016). Proof-of-concept study to assess the nociceptin receptor antagonist LY2940094 as a new treatment for alcohol dependence. Alcoholism: Clinical and Experimental Research, 40, 1935–1944. doi:10.1111/acer.13147
  • Raddad, E., Chappell, A., Meyer, J., Wilson, A., Ruegg, C. E., Tauscher, J., … Verfaille, S. J. (2016). Occupancy of Nociceptin/Orphanin FQ peptide receptors by the antagonist LY2940094 in rats and healthy human subjects. Drug Metabolism & Disposition, 44, 1536–1542. doi:10.1124/dmd.116.070359
  • Recker, M. D., & Higgins, G. A. (2004). The opioid receptor like-1 receptor agonist Ro 64-6198 (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) produces a discriminative stimulus in rats distinct from that of a mu, kappa, and delta opioid receptor agonist cue. Journal of Pharmacology and Experimental Therapeutics, 311, 652–658. doi:10.1124/jpet.104.071423
  • Rodi, D., Zucchini, S., Simonato, M., Cifani, C., Massi, M., & Polidori, C. (2008). Functional antagonism between nociceptin/orphanin FQ (N/OFQ) and corticotropin-releasing factor (CRF) in the rat brain: evidence for involvement of the bed nucleus of the stria terminalis. Psychopharmacology (Berl), 196, 523–531. doi:10.1007/s00213-007-0985-7
  • Rorick-Kehn, L. M., Ciccocioppo, R., Wong, C. J., Witkin, J. M., Martinez-Grau, M. A., Stopponi, S., … Statnick, M. A. (2016). A novel, orally bioavailable nociceptin receptor antagonist, LY2940094, reduces ethanol self-administration and ethanol seeking in animal models. Alcoholism: Clinical and Experimental Research, 40, 945–954. doi:10.1111/acer.13052
  • Saladin, M. E., Drobes, D. J., Coffey, S. F., Dansky, B. S., Brady, K. T., & Kilpatrick, D. G. (2003). PTSD symptom severity as a predictor of cue-elicited drug craving in victims of violent crime. Addictive Behaviors, 28, 1611–1629. doi:10.1016/j.addbeh.2003.08.037
  • Sledjeski, E. M., Speisman, B., & Dierker, L. C. (2008). Does number of lifetime traumas explain the relationship between PTSD and chronic medical conditions? Answers from the National Comorbidity Survey-Replication (NCS-R). Journal of Behavioral Medicine, 31, 341–349. doi:10.1007/s10865-008-9158-3
  • Smith, S. M., & Vale, W. W. (2006). The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues in Clinical Neuroscience, 8, 383–395.
  • Sra, J., Bremner, S., Krum, D., Dhala, A., Blanck, Z., Deshpande, S., … Akhtar, M. (1997). The effect of biphasic waveform tilt in transvenous atrial defibrillation. Pacing and Clinical Electrophysiology, 20, 1613–1618.
  • Stein, M. B., Simmons, A. N., Feinstein, J. S., & Paulus, M. P. (2007). Increased amygdala and insula activation during emotion processing in anxiety-prone subjects. The American Journal of Psychiatry, 164, 318–327. doi:10.1176/ajp.2007.164.2.318
  • Sullivan, G. M., Parsey, R. V., Kumar, J. S., Arango, V., Kassir, S. A., Huang, Y. Y., … Mann, J. J. (2007). PET Imaging of CRF1 with [11C]R121920 and [11C]DMP696: is the target of sufficient density?. Nuclear Medicine and Biology, 34, 353–361. doi:10.1016/j.nucmedbio.2007.01.012
  • Ulrich-Lai, Y. M., & Herman, J. P. (2009). Neural regulation of endocrine and autonomic stress responses. Nature Reviews Neuroscience, 10, 397–409. doi:10.1038/nrn2647
  • Ulrich-Lai, Y. M., Ostrander, M. M., Thomas, I. M., Packard, B. A., Furay, A. R., Dolgas, C. M., … Herman, J. P. (2007). Daily limited access to sweetened drink attenuates hypothalamic-pituitary-adrenocortical axis stress responses. Endocrinology, 148, 1823–1834. doi:10.1210/en.2006-1241
  • Vitale, G., Arletti, R., Ruggieri, V., Cifani, C., & Massi, M. (2006). Anxiolytic-like effects of nociceptin/orphanin FQ in the elevated plus maze and in the conditioned defensive burying test in rats. Peptides, 27, 2193–2200. doi:10.1016/j.peptides.2006.04.003
  • Weiss, N. H., Tull, M. T., Anestis, M. D., & Gratz, K. L. (2013). The relative and unique contributions of emotion dysregulation and impulsivity to posttraumatic stress disorder among substance dependent inpatients. Drug and Alcohol Dependence, 128, 45–51. doi:10.1016/j.drugalcdep.2012.07.017
  • Yamamoto, S., Morinobu, S., Takei, S., Fuchikami, M., Matsuki, A., Yamawaki, S., & Liberzon, I. (2009a). Single prolonged stress: toward an animal model of posttraumatic stress disorder. Depression and Anxiety, 26, 1110–1117. doi:10.1002/da.20629
  • Yamamoto, T., Miyazaki, R., & Yamada, T. (2009b). Intracerebroventricular administration of 26RFa produces an analgesic effect in the rat formalin test. Peptides, 30, 1683–1688. doi:10.1016/j.peptides.2009.05.027
  • Zhang, Y., Simpson-Durand, C. D., & Standifer, K. M. (2015). Nociceptin/orphanin FQ peptide receptor antagonist JTC-801 reverses pain and anxiety symptoms in a rat model of post-traumatic stress disorder. British Journal of Pharmacology, 172, 571–582. doi:10.1111/bph.12701

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.