211
Views
8
CrossRef citations to date
0
Altmetric
Research Article

Pharmacological Validation of Candidate Causal Sleep Genes Identified in an N2 Cross

, , , , , , , , , , , , , , , , & show all
Pages 167-181 | Received 26 Aug 2011, Accepted 26 Sep 2011, Published online: 17 Nov 2011

REFERENCES

  • Bourne, J. A. (2001). SCH 23390: The first selective dopamine D-1-like receptor antagonist. CNS Drug Rev, 7 399–414.
  • Brisbare-Roch, C., Dingemanse, J., Koberstein, R., Hoever, P., Aissaoui, H., Flores, S., . (2007). Promotion of sleep by targeting the orexin system in rats, dogs and humans. Nat Med, 13, 150–155.
  • Broman, K. W., Wu, H., Sen, S., & Churchill, G. A. (2003). R/qtl: QTL mapping in experimental crosses. Bioinformatics, 19, 889–890.
  • Caylak, E. (2009). The genetics of sleep disorders in humans: Narcolepsy, restless legs syndrome, and obstructive sleep apnea syndrome. Am J Med Genet.A, 149A, 2612–2626.
  • Chabas, D., Taheri, S., Renier, C., & Mignot, E. (2003). The genetics of narcolepsy. Annu Rev Genomics Human Genet, 4, 459–483.
  • Cirelli, C. (2009). The genetic and molecular regulation of sleep: From fruit flies to humans. Nat Rev Neurosci, 10, 549–560.
  • Cirelli, C., Gutierrez, C. M., & Tononi, G. (2004). Extensive and divergent effects of sleep and wakefulness on brain gene expression. Neuron, 41, 35–43.
  • Cohrs, S. (2008). Sleep disturbances in patients with schizophrenia impact and effect of antipsychotics. CNS Drugs, 22, 939–962.
  • Compta, Y., Santamaria, J., Ratti, L., Tolosa, E., Iranzo, A., Munoz, E., . (2009). Cerebrospinal hypocretin, daytime sleepiness and sleep architecture in Parkinson's disease dementia. Brain, 132, 3308–3317.
  • Cutrer, F. M., Yu, X. J., Ayata, G., Moskowitz, M. A., & Waeber, C. (1999). Effects of PNU-109,291, a selective 5-HT1D receptor agonist, on electrically induced dural plasma extravasation and capsaicin-evoked c-fos immunoreactivity within trigeminal nucleus caudalis. Neuropharmacology, 38, 1043–1053.
  • Dani, J. A., & Bertrand, D. (2007). Nicotinic acetylcholine receptors and nicotinic cholinergic mechanisms of the central nervous system. Annu Rev Pharmacol Toxicol, 47, 699–729.
  • Dauviltiers, Y., Maret, S., & Tafti, M. (2005). Genetics of normal and pathological sleep in humans. Sleep Med Rev, 9, 91–100.
  • Devineni, D., Skerjanec, A., & Woodworth, T. G. (2005). Low central nervous system (CNS) penetration by darifenacin, a muscarinic M3 selective receptor antagonist, in rats pA2. Online E-J Br Pharmacol Soc, 3, A092.
  • Di Fabio, R., Pellacani, A., Faedo, S., Roth, A., Piccoli, L., Gerrard, P., . (2011). Discovery process and pharmacological characterization of a novel dual orexin 1 and orexin 2 receptor antagonist useful for treatment of sleep disorders. Bioorgan Med Chem Lett, 21, 5562–5567.
  • Franken, P., & Tafti, M. (2003). Genetics of sleep and sleep disorders. Front Biosci, 8, E381–E397.
  • Gonzales, D., Rennard, S. I., Nides, M., Oncken, C., Azoulay, S., Billing, C. B., . (2006). Varenicline, an alpha 4 beta 2 nicotinic acetylcholine receptor partial agonist, vs sustained-release bupropion and placebo for smoking cessation—A randomized controlled trial. JAMA, 296, 47–55.
  • Hagan, J. J., Slade, P. D., Gaster, L., Jeffrey, P., Hatcher, J. P., & Middlemiss, D. N. (1997). Stimulation of 5-HT1B receptors causes hypothermia in the guinea pig. Eur J Pharmacol, 331, 169–174.
  • Hallmayer, J., Faraco, J., Lin, L., Hesselson, S., Winkelmann, J., Kawashima, M., . (2009). Narcolepsy is strongly associated with the T-cell receptor alpha locus. Nat Genet, 41, 708–711.
  • Hayes, M. R., Kanoski, S. E., Alhadeff, A. L., & Grill, H. J. (2011). Comparative effects of the long-acting GLP-1 receptor ligands, liraglutide and exendin-4, on food intake and body weight suppression in rats. Obesity, 19, 1342–1349.
  • Hor, H., Kutalik, Z., Dauvilliers, Y., Valsesia, A., Lammers, G. J., Donjacour, C. E. H. M., . (2010). Genome-wide association study identifies new HLA class II haplotypes strongly protective against narcolepsy. Nat Genet, 42, 786–789.
  • Jiang, P., Striz, M., Wisor, J. P., & O’Hara, B. F. (2011). Behavioral and genetic dissection of a mouse model for advanced sleep phase syndrome. SLEEP, 34, 39–48.
  • Kanoski, S. E., Fortin, S. M., Arnold, M., Grill, H. J., & Hayes, M. R. (2011). Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4. Endocrinology, 152, 3103–3112.
  • Kayser, V., Aubel, B., Hamon, M., & Bourgoin, S. (2002). The antimigraine 5-HT1B/1D receptor agonists, sumatriptan, zolmitriptan and dihydroergotamine, attenuate pain-related behaviour in a rat model of trigeminal neuropathic pain. Br J Pharmacol, 137, 1287–1297.
  • Kornum, B. R., Kawashima, M., Faraco, J., Lin, L., Rico, T. J., Hesselson, S., . (2011). Common variants in P2RY11 are associated with narcolepsy. Nat Genet, 43, 66–U90.
  • Lin, L., Faraco, J., Li, R., Kadotani, H., Rogers, W., Lin, X. Y., . (1999). The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell, 98, 365–376.
  • Mackiewicz, M., Shockley, K. R., Romer, M. A., Galante, R. J., Zimmerman, J. E., Naidoo, N., . (2007). Macromolecule biosynthesis: A key function of sleep. Physiol Genomics, 31, 441–457.
  • Mackiewicz, M., Zimmerman, J. E., Shockley, K. R., Churchill, G. A., & Pack, A. I. (2009). What are microarrays teaching us about sleep? Trends Mol Med, 15, 79–87.
  • Millstein, J., Winrow, C. J., Kasarskis, A., Owens, J. R., Zhou, L., Summa, K. C., . (2011). Identification of causal genes, networks and transcriptional regulators of REM sleep and wake. SLEEP, 34, 1469–1477.
  • Millstein, J., Zhang, B., Zhu, J., & Schadt, E. E. (2009). Disentangling molecular relationships with a causal inference test. BMC Genetics, 10, 23.
  • Missale, C., Nash, S. R., Robinson, S. W., Jaber, M., & Caron, M. G. (1998). Dopamine receptors: From structure to function. Physiol Rev, 78, 189–225.
  • Miyagawa, T., Kawashima, M., Nishida, N., Ohashi, J., Kimura, R., Fujimoto, A., . (2008). Variant between CPT1B and CHKB associated with susceptibility to narcolepsy. Nat Genet, 40, 1324–1328.
  • Nishino, S., & Okuro, M. (2010). Emerging treatments for narcolepsy and its related disorders. Expert Opin Emerg Drugs, 15, 139–158.
  • Pauwels, P. J., & John, G. W. (1999). Present and future of 5-HT receptor agonists as antimigraine drugs. Clin Neuropharmacol, 22, 123–136.
  • Peterson, M. J., & Benca, R. M. (2006). Sleep in mood disorders. Psychiatr Clin N Am, 29, 1009–1032.
  • Pinheiro, J.C., and Bates, D.M. (2000) Mixed-Effects Models in S and S-PLUS, Springer, New York, USA.
  • Renger, J. J., Dunn, S. L., Motzel, S. L., Johnson, C., & Koblan, K. S. (2004). Sub-chronic administration of zolpidem affects modifications to rat sleep architecture. Brain Res, 1010, 45–54.
  • Sajja, R. K., & Rahman, S. (2011). Lobeline and cytisine reduce voluntary ethanol drinking behavior in male C57BL/6J mice. Progress Neuro-Psychopharmacol Biol Psychiatry, 35, 257–264.
  • Saper, C. B., Scammell, T. E., & Lu, J. (2005). Hypothalamic regulation of sleep and circadian rhythms. Nature, 437, 1257–1263.
  • Schadt, E. E., Lamb, J., Yang, X., Zhu, J., Edwards, S., GuhaThakurta, D., . (2005). An integrative genomics approach to infer causal associations between gene expression and disease. Nat Genet, 37, 710–717.
  • Sehgal, A., & Mignot, E. (2011). Genetics of sleep and sleep disorders. Cell, 146, 194–207.
  • Shipe, W. D., Barrow, J. C., Yang, Z. Q., Lindsley, C. W., Yang, F. V., Schlegel, K. A., . (2008). Design, synthesis, and evaluation of a novel 4-aminomethyl-4-fluoropiperidine as a T-type Ca(2+) channel antagonist. J Med Chem, 51, 3692–3695.
  • Slassi, A., Isaac, M., & Arora, J. (2001). Novel serotonergic and non-serotonergic migraine headache therapies. Exp Opin Therapeut Patents, 11, 625–649.
  • Tafti, M. (2009). Genetic aspects of normal and disturbed sleep. Sleep Med, 10(Suppl 1), S17–S21.
  • Thompson, C. L., Wisor, J. P., Lee, C. K., Pathak, S. D., Gerashchenko, D., Smith, K. A., . (2010). Molecular and anatomical signatures of sleep deprivation in the mouse brain. Front Neurosci, 4, 165.
  • Tononi, G., & Cirelli, C. (2003). Sleep and synaptic homeostasis: A hypothesis. Brain Res Bull, 62, 143–150.
  • Uebele, V. N., Gotter, A. L., Nuss, C. E., Kraus, R. L., Doran, S. M., Garson, S. L., . (2009). Antagonism of T-type calcium channels inhibits high-fat diet-induced weight gain in mice. J Clin Invest, 119, 1659–1667.
  • Venables, W. N., & Ripley, B. D. (2002). Modern Applied Statistics with S. (4th ed.) New York: Springer.
  • Vilaro, M. T., Palacios, J. M., & Mengod, G. (1994). Multiplicity of muscarinic autoreceptor subtypes—Comparison of the distribution of cholinergic cells and cells containing messenger-RNA for 5 subtypes of muscarinic receptors in the rat-brain. Mol Brain Res, 21, 30–46.
  • Whitman, D. B., Cox, C. D., Breslin, M. J., Brashear, K. M., Schreier, J. D., Bogusky, M. J., . (2009). Discovery of a potent, CNS-penetrant orexin receptor antagonist based on an N,N-disubstituted-1,4-diazepane scaffold that promotes sleep in rats. Chemmedchem, 4, 1069–1074.
  • Winrow, C. J., Gotter, A. L., Cox, C. D., Doran, S. M., Tannenbaum, P. L., Breslin, M. J., . (2011). Promotion of sleep by suvorexant—A novel dual orexin receptor antagonist. J Neurogenet, 25, 52–61.
  • Winrow, C. J., Williams, D. L., Kasarskis, A., Millstein, J., Laposky, A. D., Yang, H. S., . (2009). Uncovering the genetic landscape for multiple sleep-wake traits. PLoS One, 4, e5161.
  • Yin, X. P., Wei, D. Z., Yi, L. N., Tao, X. Y., & Ma, Y. S. (2005). Expression and purification of exendin-4, a GLP-1 receptor agonist, in Escherichia coli. Prot Express Purif, 41, 259–265.
  • Zaniewska, M., McCreary, A. C., Stefanski, R., Przegalinski, E., & Filip, M. (2008). Effect of varenicline on the acute and repeated locomotor responses to nicotine in rats. Synapse, 62, 935–939.
  • Zee, P. C., & Turek, F. W. (2006). Sleep and health—Everywhere and in both directions. Arch Intern Med, 166, 1686–1688.
  • Zhang, C., Fang, Y. R., & Li, M. (2011). Olanzapine and risperidone disrupt conditioned avoidance responding by selectively weakening motivational salience of conditioned stimulus: Further evidence. Pharmacol Biochem Behav, 98, 155–160.

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.