Potential Utility of Biased GPCR Signaling for Treatment of Psychiatric Disorders
Abstract
1. Introduction
2. Canonical and Noncanonical GPCR Signaling Pathways
3. Modes of Action of Antipsychotics in Psychiatric Disorders
3.1. Aripiprazole
3.2. Quetiapine
4. Elucidation of D2R-Mediated Biased Functions by Genetically Engineered Biased D2R Mutants
5. Potential Biased Antipsychotics
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Gandal, M.J.; Haney, J.R.; Parikshak, N.N.; Leppa, V.; Ramaswami, G.; Hartl, C.; Schork, A.J.; Appadurai, V.; Buil, A.; Werge, T.M.; et al. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science 2018, 359, 693–697. [Google Scholar] [CrossRef] [PubMed]
- Teroganova, N.; Girshkin, L.; Suter, C.M.; Green, M.J. DNA methylation in peripheral tissue of schizophrenia and bipolar disorder: A systematic review. BMC Genet. 2016, 17, 27. [Google Scholar] [CrossRef] [PubMed]
- Hopkins, A.L. Network pharmacology: The next paradigm in drug discovery. Nat. Chem. Biol. 2008, 4, 682–690. [Google Scholar] [CrossRef]
- Masri, B.; Salahpour, A.; Didriksen, M.; Ghisi, V.; Beaulieu, J.M.; Gainetdinov, R.R.; Caron, M.G. Antagonism of dopamine D2 receptor/beta-arrestin 2 interaction is a common property of clinically effective antipsychotics. Proc. Natl. Acad. Sci. USA 2008, 105, 13656–13661. [Google Scholar] [CrossRef] [PubMed]
- Khan, S.M.; Sung, J.Y.; Hebert, T.E. Gbetagamma subunits-Different spaces, different faces. Pharm. Res. 2016, 111, 434–441. [Google Scholar] [CrossRef] [PubMed]
- Bologna, Z.; Teoh, J.P.; Bayoumi, A.S.; Tang, Y.; Kim, I.M. Biased G Protein-Coupled Receptor Signaling: New Player in Modulating Physiology and Pathology. Biomol. Ther. 2017, 25, 12–25. [Google Scholar] [CrossRef] [PubMed]
- Greengard, P. The neurobiology of slow synaptic transmission. Science 2001, 294, 1024–1030. [Google Scholar] [CrossRef] [PubMed]
- Grundmann, M.; Merten, N.; Malfacini, D.; Inoue, A.; Preis, P.; Simon, K.; Ruttiger, N.; Ziegler, N.; Benkel, T.; Schmitt, N.K.; et al. Lack of beta-arrestin signaling in the absence of active G proteins. Nat. Commun. 2018, 9, 341. [Google Scholar] [CrossRef]
- O’Hayre, M.; Eichel, K.; Avino, S.; Zhao, X.; Steffen, D.J.; Feng, X.; Kawakami, K.; Aoki, J.; Messer, K.; Sunahara, R.; et al. Genetic evidence that beta-arrestins are dispensable for the initiation of beta2-adrenergic receptor signaling to ERK. Sci. Signal. 2017, 10, eaal3395. [Google Scholar] [CrossRef]
- Salahpour, A.; Espinoza, S.; Masri, B.; Lam, V.; Barak, L.S.; Gainetdinov, R.R. BRET biosensors to study GPCR biology, pharmacology, and signal transduction. Front. Endocrinol. 2012, 3, 105. [Google Scholar] [CrossRef]
- Howes, O.D.; Kapur, S. The dopamine hypothesis of schizophrenia: Version III—The final common pathway. Schizophr. Bull. 2009, 35, 549–562. [Google Scholar] [CrossRef] [PubMed]
- Bernheimer, H.; Birkmayer, W.; Hornykiewicz, O.; Jellinger, K.; Seitelberger, F. Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J. Neurol. Sci. 1973, 20, 415–455. [Google Scholar] [CrossRef]
- Pauls, D.L.; Abramovitch, A.; Rauch, S.L.; Geller, D.A. Obsessive-compulsive disorder: An integrative genetic and neurobiological perspective. Nat. Rev. Neurosci. 2014, 15, 410–424. [Google Scholar] [CrossRef] [PubMed]
- De Bartolomeis, A.; Buonaguro, E.F.; Iasevoli, F.; Tomasetti, C. The emerging role of dopamine-glutamate interaction and of the postsynaptic density in bipolar disorder pathophysiology: Implications for treatment. J. Psychopharmacol. 2014, 28, 505–526. [Google Scholar] [CrossRef] [PubMed]
- Creese, I.; Burt, D.R.; Snyder, S.H. Dopamine receptor binding predicts clinical and pharmacological potencies of antischizophrenic drugs. Science 1976, 192, 481–483. [Google Scholar] [CrossRef]
- Lieberman, J.A.; Kane, J.M.; Gadaleta, D.; Brenner, R.; Lesser, M.S.; Kinon, B. Methylphenidate challenge as a predictor of relapse in schizophrenia. Am. J. Psychiatry 1984, 141, 633–638. [Google Scholar] [PubMed]
- Davidson, M.; Keefe, R.S.; Mohs, R.C.; Siever, L.J.; Losonczy, M.F.; Horvath, T.B.; Davis, K.L. L-dopa challenge and relapse in schizophrenia. Am. J. Psychiatry 1987, 144, 934–938. [Google Scholar]
- Missale, C.; Nash, S.R.; Robinson, S.W.; Jaber, M.; Caron, M.G. Dopamine receptors: From structure to function. Physiol. Rev. 1998, 78, 189–225. [Google Scholar] [CrossRef]
- Surmeier, D.J.; Ding, J.; Day, M.; Wang, Z.; Shen, W. D1 and D2 dopamine-receptor modulation of striatal glutamatergic signaling in striatal medium spiny neurons. Trends Neurosci. 2007, 30, 228–235. [Google Scholar] [CrossRef]
- Bolam, J.P.; Hanley, J.J.; Booth, P.A.; Bevan, M.D. Synaptic organisation of the basal ganglia. J. Anat. 2000, 196 Pt 4, 527–542. [Google Scholar] [CrossRef]
- Ena, S.; de Kerchove d’Exaerde, A.; Schiffmann, S.N. Unraveling the differential functions and regulation of striatal neuron sub-populations in motor control, reward, and motivational processes. Front. Behav. Neurosci. 2011, 5, 47. [Google Scholar] [CrossRef] [PubMed]
- Cachope, R.; Cheer, J.F. Local control of striatal dopamine release. Front. Behav. Neurosci. 2014, 8, 188. [Google Scholar] [CrossRef] [PubMed]
- Kawaguchi, Y.; Wilson, C.J.; Augood, S.J.; Emson, P.C. Striatal interneurones: Chemical, physiological and morphological characterization. Trends Neurosci. 1995, 18, 527–535. [Google Scholar] [CrossRef]
- Tepper, J.M.; Bolam, J.P. Functional diversity and specificity of neostriatal interneurons. Curr. Opin. Neurobiol. 2004, 14, 685–692. [Google Scholar] [CrossRef] [PubMed]
- Graybiel, A.M.; Canales, J.J.; Capper-Loup, C. Levodopa-induced dyskinesias and dopamine-dependent stereotypies: A new hypothesis. Trends Neurosci. 2000, 23 (Suppl. 10), S71–S77. [Google Scholar] [CrossRef]
- Gerfen, C.R.; Young, W.S., 3rd. Distribution of striatonigral and striatopallidal peptidergic neurons in both patch and matrix compartments: An in situ hybridization histochemistry and fluorescent retrograde tracing study. Brain Res. 1988, 460, 161–167. [Google Scholar] [CrossRef]
- Gerfen, C.R.; Engber, T.M.; Mahan, L.C.; Susel, Z.; Chase, T.N.; Monsma, F.J., Jr.; Sibley, D.R. D1 and D2 dopamine receptor-regulated gene expression of striatonigral and striatopallidal neurons. Science 1990, 250, 1429–1432. [Google Scholar] [CrossRef] [PubMed]
- Komatsu, H.; Maruyama, M.; Yao, S.; Shinohara, T.; Sakuma, K.; Imaichi, S.; Chikatsu, T.; Kuniyeda, K.; Siu, F.K.; Peng, L.S.; et al. Anatomical transcriptome of G protein-coupled receptors leads to the identification of a novel therapeutic candidate GPR52 for psychiatric disorders. PLoS ONE 2014, 9, e90134. [Google Scholar] [CrossRef] [PubMed]
- Schiffmann, S.N.; Jacobs, O.; Vanderhaeghen, J.J. Striatal restricted adenosine A2 receptor (RDC8) is expressed by enkephalin but not by substance P neurons: An in situ hybridization histochemistry study. J. Neurochem. 1991, 57, 1062–1067. [Google Scholar] [CrossRef]
- Schiffmann, S.N.; Fisone, G.; Moresco, R.; Cunha, R.A.; Ferre, S. Adenosine A2A receptors and basal ganglia physiology. Prog. Neurobiol. 2007, 83, 277–292. [Google Scholar] [CrossRef]
- Lobo, M.K.; Cui, Y.; Ostlund, S.B.; Balleine, B.W.; Yang, X.W. Genetic control of instrumental conditioning by striatopallidal neuron-specific S1P receptor Gpr6. Nat. Neurosci. 2007, 10, 1395–1397. [Google Scholar] [CrossRef] [PubMed]
- Quintana, A.; Sanz, E.; Wang, W.; Storey, G.P.; Guler, A.D.; Wanat, M.J.; Roller, B.A.; La Torre, A.; Amieux, P.S.; McKnight, G.S.; et al. Lack of GPR88 enhances medium spiny neuron activity and alters motor- and cue-dependent behaviors. Nat. Neurosci. 2012, 15, 1547–1555. [Google Scholar] [CrossRef] [PubMed]
- Albin, R.L.; Young, A.B.; Penney, J.B. The functional anatomy of basal ganglia disorders. Trends Neurosci. 1989, 12, 366–375. [Google Scholar] [CrossRef]
- DeLong, M.R.; Wichmann, T. Circuits and circuit disorders of the basal ganglia. Arch. Neurol. 2007, 64, 20–24. [Google Scholar] [CrossRef] [PubMed]
- Svenningsson, P.; Tzavara, E.T.; Carruthers, R.; Rachleff, I.; Wattler, S.; Nehls, M.; McKinzie, D.L.; Fienberg, A.A.; Nomikos, G.G.; Greengard, P. Diverse psychotomimetics act through a common signaling pathway. Science 2003, 302, 1412–1415. [Google Scholar] [CrossRef] [PubMed]
- Bateup, H.S.; Santini, E.; Shen, W.; Birnbaum, S.; Valjent, E.; Surmeier, D.J.; Fisone, G.; Nestler, E.J.; Greengard, P. Distinct subclasses of medium spiny neurons differentially regulate striatal motor behaviors. Proc. Natl. Acad. Sci. USA 2010, 107, 14845–14850. [Google Scholar] [CrossRef] [PubMed]
- Nishiyama, K.; Suzuki, H.; Harasawa, T.; Suzuki, N.; Kurimoto, E.; Kawai, T.; Maruyama, M.; Komatsu, H.; Sakuma, K.; Shimizu, Y.; et al. FTBMT, a Novel and Selective GPR52 Agonist, Demonstrates Antipsychotic-Like and Procognitive Effects in Rodents, Revealing a Potential Therapeutic Agent for Schizophrenia. J. Pharm. Exp. 2017, 363, 253–264. [Google Scholar] [CrossRef]
- Nishiyama, K.; Suzuki, H.; Maruyama, M.; Yoshihara, T.; Ohta, H. Genetic deletion of GPR52 enhances the locomotor-stimulating effect of an adenosine A2A receptor antagonist in mice: A potential role of GPR52 in the function of striatopallidal neurons. Brain Res. 2017, 1670, 24–31. [Google Scholar] [CrossRef]
- Dudman, J.T.; Eaton, M.E.; Rajadhyaksha, A.; Macias, W.; Taher, M.; Barczak, A.; Kameyama, K.; Huganir, R.; Konradi, C. Dopamine D1 receptors mediate CREB phosphorylation via phosphorylation of the NMDA receptor at Ser897-NR1. J. Neurochem. 2003, 87, 922–934. [Google Scholar] [CrossRef]
- Chen, G.; Greengard, P.; Yan, Z. Potentiation of NMDA receptor currents by dopamine D1 receptors in prefrontal cortex. Proc. Natl. Acad. Sci. USA 2004, 101, 2596–2600. [Google Scholar] [CrossRef]
- Gainetdinov, R.R.; Premont, R.T.; Bohn, L.M.; Lefkowitz, R.J.; Caron, M.G. Desensitization of G protein-coupled receptors and neuronal functions. Annu. Rev. Neurosci. 2004, 27, 107–144. [Google Scholar] [CrossRef] [PubMed]
- Beaulieu, J.M.; Sotnikova, T.D.; Marion, S.; Lefkowitz, R.J.; Gainetdinov, R.R.; Caron, M.G. An Akt/beta-arrestin 2/PP2A signaling complex mediates dopaminergic neurotransmission and behavior. Cell 2005, 122, 261–273. [Google Scholar] [CrossRef] [PubMed]
- Beaulieu, J.M.; Marion, S.; Rodriguiz, R.M.; Medvedev, I.O.; Sotnikova, T.D.; Ghisi, V.; Wetsel, W.C.; Lefkowitz, R.J.; Gainetdinov, R.R.; Caron, M.G. A beta-arrestin 2 signaling complex mediates lithium action on behavior. Cell 2008, 132, 125–136. [Google Scholar] [CrossRef] [PubMed]
- Beaulieu, J.M.; Tirotta, E.; Sotnikova, T.D.; Masri, B.; Salahpour, A.; Gainetdinov, R.R.; Borrelli, E.; Caron, M.G. Regulation of Akt signaling by D2 and D3 dopamine receptors in vivo. J. Neurosci. 2007, 27, 881–885. [Google Scholar] [CrossRef] [PubMed]
- Urs, N.M.; Snyder, J.C.; Jacobsen, J.P.; Peterson, S.M.; Caron, M.G. Deletion of GSK3beta in D2R-expressing neurons reveals distinct roles for beta-arrestin signaling in antipsychotic and lithium action. Proc. Natl. Acad. Sci. USA 2012, 109, 20732–20737. [Google Scholar] [CrossRef]
- Su, P.; Li, S.; Chen, S.; Lipina, T.V.; Wang, M.; Lai, T.K.; Lee, F.H.; Zhang, H.; Zhai, D.; Ferguson, S.S.; et al. A dopamine D2 receptor-DISC1 protein complex may contribute to antipsychotic-like effects. Neuron 2014, 84, 1302–1316. [Google Scholar] [CrossRef] [PubMed]
- Yatham, L.N.; Kennedy, S.H.; Parikh, S.V.; Schaffer, A.; Bond, D.J.; Frey, B.N.; Sharma, V.; Goldstein, B.I.; Rej, S.; Beaulieu, S.; et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) and International Society for Bipolar Disorders (ISBD) 2018 guidelines for the management of patients with bipolar disorder. Bipolar Disord. 2018, 20, 97–170. [Google Scholar] [CrossRef]
- Ripke, S.; Neale, B.M.; Corvin, A.; Walters, J.T.; Farh, K.H.; Holmans, P.A.; Pers, T.H. Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014, 511, 421–427. [Google Scholar]
- Kaneko, K. Negative Symptoms and Cognitive Impairments in Schizophrenia: Two Key Symptoms Negatively Influencing Social Functioning. Yonago Acta Med. 2018, 61, 91–102. [Google Scholar] [CrossRef]
- Meltzer, H.Y.; Matsubara, S.; Lee, J.C. Classification of typical and atypical antipsychotic drugs on the basis of dopamine D-1, D-2 and serotonin2 pKi values. J. Pharm. Exp. 1989, 251, 238–246. [Google Scholar]
- Abi-Dargham, A.; Laruelle, M. Mechanisms of action of second generation antipsychotic drugs in schizophrenia: Insights from brain imaging studies. Eur. Psychiatry 2005, 20, 15–27. [Google Scholar] [CrossRef] [PubMed]
- Davis, K.L.; Kahn, R.S.; Ko, G.; Davidson, M. Dopamine in schizophrenia: A review and reconceptualization. Am. J. Psychiatry 1991, 148, 1474–1486. [Google Scholar] [PubMed]
- Pycock, C.J.; Kerwin, R.W.; Carter, C.J. Effect of lesion of cortical dopamine terminals on subcortical dopamine receptors in rats. Nature 1980, 286, 74–76. [Google Scholar] [CrossRef] [PubMed]
- Weinberger, D.R. Implications of normal brain development for the pathogenesis of schizophrenia. Arch. Gen. Psychiatry 1987, 44, 660–669. [Google Scholar] [CrossRef] [PubMed]
- Beaulieu, J.M.; Sotnikova, T.D.; Yao, W.D.; Kockeritz, L.; Woodgett, J.R.; Gainetdinov, R.R.; Caron, M.G. Lithium antagonizes dopamine-dependent behaviors mediated by an AKT/glycogen synthase kinase 3 signaling cascade. Proc. Natl. Acad. Sci. USA 2004, 101, 5099–5104. [Google Scholar] [CrossRef] [PubMed]
- Kim, I.M.; Tilley, D.G.; Chen, J.; Salazar, N.C.; Whalen, E.J.; Violin, J.D.; Rockman, H.A. Beta-blockers alprenolol and carvedilol stimulate beta-arrestin-mediated EGFR transactivation. Proc. Natl. Acad. Sci. USA 2008, 105, 14555–14560. [Google Scholar] [CrossRef] [PubMed]
- Urs, N.M.; Gee, S.M.; Pack, T.F.; McCorvy, J.D.; Evron, T.; Snyder, J.C.; Yang, X.; Rodriguiz, R.M.; Borrelli, E.; Wetsel, W.C.; et al. Distinct cortical and striatal actions of a beta-arrestin-biased dopamine D2 receptor ligand reveal unique antipsychotic-like properties. Proc. Natl. Acad. Sci. USA 2016, 113, E8178–E8186. [Google Scholar] [CrossRef]
- Emamian, E.S.; Hall, D.; Birnbaum, M.J.; Karayiorgou, M.; Gogos, J.A. Convergent evidence for impaired AKT1-GSK3beta signaling in schizophrenia. Nat. Genet. 2004, 36, 131–137. [Google Scholar] [CrossRef]
- Weiwer, M.; Xu, Q.; Gale, J.P.; Lewis, M.; Campbell, A.J.; Schroeder, F.A.; Van de Bittner, G.C.; Walk, M.; Amaya, A.; Su, P.; et al. Functionally Biased D2R Antagonists: Targeting the beta-Arrestin Pathway to Improve Antipsychotic Treatment. ACS Chem. Biol. 2018, 13, 1038–1047. [Google Scholar] [CrossRef]
- Muneer, A. The Treatment of Adult Bipolar Disorder with Aripiprazole: A Systematic Review. Cureus 2016, 8, e562. [Google Scholar] [CrossRef]
- Ozaki, N.; Otsubo, T.; Kato, M.; Higuchi, T.; Ono, H.; Kamijima, K. Efficacy of aripiprazole augmentation in Japanese patients with major depressive disorder: A subgroup analysis and Montgomery-Asberg Depression Rating Scale and Hamilton Rating Scale for Depression item analyses of the Aripiprazole Depression Multicenter Efficacy study. Psychiatry Clin. Neurosci. 2015, 69, 34–42. [Google Scholar] [PubMed]
- Sonnenschein, S.F.; Gill, K.M.; Grace, A.A. State-dependent effects of the D2 partial agonist aripiprazole on dopamine neuron activity in the MAM neurodevelopmental model of schizophrenia. Neuropsychopharmacology 2019, 44, 572–580. [Google Scholar] [CrossRef] [PubMed]
- Ma, G.F.; Raivio, N.; Sabria, J.; Ortiz, J. Agonist and antagonist effects of aripiprazole on D(2)-like receptors controlling rat brain dopamine synthesis depend on the dopaminergic tone. Int. J. Neuropsychopharmacol. 2014, 18, pyu046. [Google Scholar] [CrossRef] [PubMed]
- Urban, J.D.; Clarke, W.P.; von Zastrow, M.; Nichols, D.E.; Kobilka, B.; Weinstein, H.; Javitch, J.A.; Roth, B.L.; Christopoulos, A.; Sexton, P.M.; et al. Functional selectivity and classical concepts of quantitative pharmacology. J. Pharm. Exp. 2007, 320, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Stahl, S.M. Dopamine system stabilizers, aripiprazole, and the next generation of antipsychotics, part 2: Illustrating their mechanism of action. J. Clin. Psychiatry 2001, 62, 923–924. [Google Scholar] [CrossRef]
- Akbarian, S.; Kim, J.J.; Potkin, S.G.; Hagman, J.O.; Tafazzoli, A.; Bunney, W.E., Jr.; Jones, E.G. Gene expression for glutamic acid decarboxylase is reduced without loss of neurons in prefrontal cortex of schizophrenics. Arch. Gen. Psychiatry 1995, 52, 258–266. [Google Scholar] [CrossRef] [PubMed]
- Thompson, M.; Weickert, C.S.; Wyatt, E.; Webster, M.J. Decreased glutamic acid decarboxylase(67) mRNA expression in multiple brain areas of patients with schizophrenia and mood disorders. J. Psychiatr. Res. 2009, 43, 970–977. [Google Scholar] [CrossRef]
- Volk, D.W.; Austin, M.C.; Pierri, J.N.; Sampson, A.R.; Lewis, D.A. Decreased glutamic acid decarboxylase67 messenger RNA expression in a subset of prefrontal cortical gamma-aminobutyric acid neurons in subjects with schizophrenia. Arch. Gen. Psychiatry 2000, 57, 237–245. [Google Scholar] [CrossRef]
- Fung, S.J.; Webster, M.J.; Sivagnanasundaram, S.; Duncan, C.; Elashoff, M.; Weickert, C.S. Expression of interneuron markers in the dorsolateral prefrontal cortex of the developing human and in schizophrenia. Am. J. Psychiatry 2010, 167, 1479–1488. [Google Scholar] [CrossRef]
- Hashimoto, T.; Bazmi, H.H.; Mirnics, K.; Wu, Q.; Sampson, A.R.; Lewis, D.A. Conserved regional patterns of GABA-related transcript expression in the neocortex of subjects with schizophrenia. Am. J. Psychiatry 2008, 165, 479–489. [Google Scholar] [CrossRef]
- Nakazawa, K.; Zsiros, V.; Jiang, Z.; Nakao, K.; Kolata, S.; Zhang, S.; Belforte, J.E. GABAergic interneuron origin of schizophrenia pathophysiology. Neuropharmacology 2012, 62, 1574–1583. [Google Scholar] [CrossRef] [PubMed]
- Lewis, D.A.; Curley, A.A.; Glausier, J.R.; Volk, D.W. Cortical parvalbumin interneurons and cognitive dysfunction in schizophrenia. Trends Neurosci. 2012, 35, 57–67. [Google Scholar] [CrossRef] [PubMed]
- Sohal, V.S.; Zhang, F.; Yizhar, O.; Deisseroth, K. Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature 2009, 459, 698–702. [Google Scholar] [CrossRef] [PubMed]
- Cho, K.K.; Hoch, R.; Lee, A.T.; Patel, T.; Rubenstein, J.L.; Sohal, V.S. Gamma rhythms link prefrontal interneuron dysfunction with cognitive inflexibility in Dlx5/6(+/-) mice. Neuron 2015, 85, 1332–1343. [Google Scholar] [CrossRef] [PubMed]
- Gefvert, O.; Bergstrom, M.; Langstrom, B.; Lundberg, T.; Lindstrom, L.; Yates, R. Time course of central nervous dopamine-D2 and 5-HT2 receptor blockade and plasma drug concentrations after discontinuation of quetiapine (Seroquel) in patients with schizophrenia. Psychopharmacology 1998, 135, 119–126. [Google Scholar] [CrossRef] [PubMed]
- Saller, C.F.; Salama, A.I. Seroquel: Biochemical profile of a potential atypical antipsychotic. Psychopharmacology 1993, 112, 285–292. [Google Scholar] [CrossRef]
- Jeong, J.H.; Bahk, W.M.; Woo, Y.S.; Lee, J.G.; Kim, M.D.; Sohn, I.; Shim, S.H.; Jon, D.I.; Seo, J.S.; Kim, W.; et al. Korean Medication Algorithm for Bipolar Disorder 2018: Comparisons with Other Treatment Guidelines. Clin. Psychopharmacol. Neurosci. 2019, 17, 155–169. [Google Scholar] [CrossRef]
- Jensen, N.H.; Rodriguiz, R.M.; Caron, M.G.; Wetsel, W.C.; Rothman, R.B.; Roth, B.L. N-desalkylquetiapine, a potent norepinephrine reuptake inhibitor and partial 5-HT1A agonist, as a putative mediator of quetiapine’s antidepressant activity. Neuropsychopharmacology 2008, 33, 2303–2312. [Google Scholar] [CrossRef]
- Calabrese, J.R.; Keck, P.E., Jr.; Macfadden, W.; Minkwitz, M.; Ketter, T.A.; Weisler, R.H.; Cutler, A.J.; McCoy, R.; Wilson, E.; Mullen, J. A randomized, double-blind, placebo-controlled trial of quetiapine in the treatment of bipolar I or II depression. Am. J. Psychiatry 2005, 162, 1351–1360. [Google Scholar] [CrossRef]
- McElroy, S.L.; Weisler, R.H.; Chang, W.; Olausson, B.; Paulsson, B.; Brecher, M.; Agambaram, V.; Merideth, C.; Nordenhem, A.; Young, A.H. A double-blind, placebo-controlled study of quetiapine and paroxetine as monotherapy in adults with bipolar depression (EMBOLDEN II). J. Clin. Psychiatry 2010, 71, 163–174. [Google Scholar] [CrossRef]
- Suppes, T.; Datto, C.; Minkwitz, M.; Nordenhem, A.; Walker, C.; Darko, D. Effectiveness of the extended release formulation of quetiapine as monotherapy for the treatment of acute bipolar depression. J. Affect. Disord. 2010, 121, 106–115. [Google Scholar] [CrossRef] [PubMed]
- Thase, M.E.; Macfadden, W.; Weisler, R.H.; Chang, W.; Paulsson, B.; Khan, A.; Calabrese, J.R. Efficacy of quetiapine monotherapy in bipolar I and II depression: A double-blind, placebo-controlled study (the BOLDER II study). J. Clin. Psychopharmacol. 2006, 26, 600–609. [Google Scholar] [CrossRef] [PubMed]
- Young, A.H.; McElroy, S.L.; Bauer, M.; Philips, N.; Chang, W.; Olausson, B.; Paulsson, B.; Brecher, M. A double-blind, placebo-controlled study of quetiapine and lithium monotherapy in adults in the acute phase of bipolar depression (EMBOLDEN I). J. Clin. Psychiatry 2010, 71, 150–162. [Google Scholar] [CrossRef] [PubMed]
- Kao, Y.C.; Ko, C.Y.; Wang, S.C.; Liu, Y.P. Protective Effects of Quetiapine on Metabolic and Inflammatory Abnormalities in Schizophrenic Patients during Exacerbated Stage. Chin. J. Physiol. 2016, 59, 69–77. [Google Scholar] [CrossRef] [PubMed]
- Wang, K.; Song, F.; Wang, H.; Wang, J.H.; Sun, Y. Quetiapine Attenuates the Neuroinflammation and Executive Function Deficit in Streptozotocin-Induced Diabetic Mice. Mediat. Inflamm. 2019, 2019, 1236082. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Zhu, S.; Wang, H.; He, J.; Zhang, Y.; Adilijiang, A.; Zhang, H.; Hartle, K.; Guo, H.; Kong, J.; et al. Astrocyte-dependent protective effect of quetiapine on GABAergic neuron is associated with the prevention of anxiety-like behaviors in aging mice after long-term treatment. J. Neurochem. 2014, 130, 780–789. [Google Scholar] [CrossRef] [PubMed]
- Luo, G.; Huang, Y.; Jia, B.; Zhang, X.; Mo, D.; Ma, N.; Gao, F.; Song, L.; Wang, B.; Miao, Z. Quetiapine prevents Abeta25-35-induced cell death in cultured neuron by enhancing brain-derived neurotrophic factor release from astrocyte. Neuroreport 2018, 29, 92–98. [Google Scholar] [CrossRef]
- Pereira, A.; Zhang, B.; Malcolm, P.; Sugiharto-Winarno, A.; Sundram, S. Quetiapine and aripiprazole signal differently to ERK, p90RSK and c-Fos in mouse frontal cortex and striatum: Role of the EGF receptor. BMC Neurosci. 2014, 15, 30. [Google Scholar] [CrossRef]
- Bui, K.; Earley, W.; Nyberg, S. Pharmacokinetic profile of the extended-release formulation of quetiapine fumarate (quetiapine XR): Clinical implications. Curr. Med. Res. Opin. 2013, 29, 813–825. [Google Scholar] [CrossRef]
- Aringhieri, S.; Kolachalam, S.; Gerace, C.; Carli, M.; Verdesca, V.; Brunacci, M.G.; Rossi, C.; Ippolito, C.; Solini, A.; Corsini, G.U.; et al. Clozapine as the most efficacious antipsychotic for activating ERK 1/2 kinases: Role of 5-HT2A receptor agonism. Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol. 2017, 27, 383–398. [Google Scholar] [CrossRef]
- Dobbs, L.K.; Kaplan, A.R.; Lemos, J.C.; Matsui, A.; Rubinstein, M.; Alvarez, V.A. Dopamine Regulation of Lateral Inhibition between Striatal Neurons Gates the Stimulant Actions of Cocaine. Neuron 2016, 90, 1100–1113. [Google Scholar] [CrossRef] [PubMed]
- Kharkwal, G.; Radl, D.; Lewis, R.; Borrelli, E. Dopamine D2 receptors in striatal output neurons enable the psychomotor effects of cocaine. Proc. Natl. Acad. Sci. USA 2016, 113, 11609–11614. [Google Scholar] [CrossRef] [PubMed]
- Lemos, J.C.; Friend, D.M.; Kaplan, A.R.; Shin, J.H.; Rubinstein, M.; Kravitz, A.V.; Alvarez, V.A. Enhanced GABA Transmission Drives Bradykinesia Following Loss of Dopamine D2 Receptor Signaling. Neuron 2016, 90, 824–838. [Google Scholar] [CrossRef] [PubMed]
- Baik, J.H.; Picetti, R.; Saiardi, A.; Thiriet, G.; Dierich, A.; Depaulis, A.; Le Meur, M.; Borrelli, E. Parkinsonian-like locomotor impairment in mice lacking dopamine D2 receptors. Nature 1995, 377, 424–428. [Google Scholar] [CrossRef] [PubMed]
- Peterson, S.M.; Pack, T.F.; Wilkins, A.D.; Urs, N.M.; Urban, D.J.; Bass, C.E.; Lichtarge, O.; Caron, M.G. Elucidation of G-protein and beta-arrestin functional selectivity at the dopamine D2 receptor. Proc. Natl. Acad. Sci. USA 2015, 112, 7097–7102. [Google Scholar] [CrossRef] [PubMed]
- Rose, S.J.; Pack, T.F.; Peterson, S.M.; Payne, K.; Borrelli, E.; Caron, M.G. Engineered D2R Variants Reveal the Balanced and Biased Contributions of G-Protein and beta-Arrestin to Dopamine-Dependent Functions. Neuropsychopharmacology 2018, 43, 1164–1173. [Google Scholar] [CrossRef] [PubMed]
- Bertran-Gonzalez, J.; Bosch, C.; Maroteaux, M.; Matamales, M.; Herve, D.; Valjent, E.; Girault, J.A. Opposing patterns of signaling activation in dopamine D1 and D2 receptor-expressing striatal neurons in response to cocaine and haloperidol. J. Neurosci. 2008, 28, 5671–5685. [Google Scholar] [CrossRef] [PubMed]
- Bonito-Oliva, A.; Pallottino, S.; Bertran-Gonzalez, J.; Girault, J.A.; Valjent, E.; Fisone, G. Haloperidol promotes mTORC1-dependent phosphorylation of ribosomal protein S6 via dopamine- and cAMP-regulated phosphoprotein of 32 kDa and inhibition of protein phosphatase-1. Neuropharmacology 2013, 72, 197–203. [Google Scholar] [CrossRef] [PubMed]
- Bertran-Gonzalez, J.; Hakansson, K.; Borgkvist, A.; Irinopoulou, T.; Brami-Cherrier, K.; Usiello, A.; Greengard, P.; Herve, D.; Girault, J.A.; Valjent, E.; et al. Histone H3 phosphorylation is under the opposite tonic control of dopamine D2 and adenosine A2A receptors in striatopallidal neurons. Neuropsychopharmacology 2009, 34, 1710–1720. [Google Scholar] [CrossRef]
- Valjent, E.; Bertran-Gonzalez, J.; Bowling, H.; Lopez, S.; Santini, E.; Matamales, M.; Bonito-Oliva, A.; Herve, D.; Hoeffer, C.; Klann, E.; et al. Haloperidol regulates the state of phosphorylation of ribosomal protein S6 via activation of PKA and phosphorylation of DARPP-32. Neuropsychopharmacology 2011, 36, 2561–2570. [Google Scholar] [CrossRef]
- Allen, J.A.; Yost, J.M.; Setola, V.; Chen, X.; Sassano, M.F.; Chen, M.; Peterson, S.; Yadav, P.N.; Huang, X.P.; Feng, B.; et al. Discovery of beta-arrestin-biased dopamine D2 ligands for probing signal transduction pathways essential for antipsychotic efficacy. Proc. Natl. Acad. Sci. USA 2011, 108, 18488–18493. [Google Scholar] [CrossRef] [PubMed]
- Donthamsetti, P.; Gallo, E.F.; Buck, D.C.; Stahl, E.L.; Zhu, Y.; Lane, J.R.; Bohn, L.M.; Neve, K.A.; Kellendonk, C.; Javitch, J.A. Arrestin recruitment to dopamine D2 receptor mediates locomotion but not incentive motivation. Mol. Psychiatry 2018. [Google Scholar] [CrossRef] [PubMed]
- Lawler, C.P.; Prioleau, C.; Lewis, M.M.; Mak, C.; Jiang, D.; Schetz, J.A.; Gonzalez, A.M.; Sibley, D.R.; Mailman, R.B. Interactions of the novel antipsychotic aripiprazole (OPC-14597) with dopamine and serotonin receptor subtypes. Neuropsychopharmacology 1999, 20, 612–627. [Google Scholar] [CrossRef]
- Sahlholm, K.; Gomez-Soler, M.; Valle-Leon, M.; Lopez-Cano, M.; Taura, J.J.; Ciruela, F.; Fernandez-Duenas, V. Antipsychotic-Like Efficacy of Dopamine D2 Receptor-Biased Ligands is Dependent on Adenosine A2A Receptor Expression. Mol. Neurobiol. 2018, 55, 4952–4958. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.F.; Moratalla, R.; Impagnatiello, F.; Grandy, D.K.; Cuellar, B.; Rubinstein, M.; Beilstein, M.A.; Hackett, E.; Fink, J.S.; Low, M.J.; et al. The role of the D(2) dopamine receptor (D(2)R) in A(2A) adenosine receptor (A(2A)R)-mediated behavioral and cellular responses as revealed by A(2A) and D(2) receptor knockout mice. Proc. Natl. Acad. Sci. USA 2001, 98, 1970–1975. [Google Scholar] [CrossRef]
- Fukuchi, M.; Tabuchi, A.; Kuwana, Y.; Watanabe, S.; Inoue, M.; Takasaki, I.; Izumi, H.; Tanaka, A.; Inoue, R.; Mori, H.; et al. Neuromodulatory effect of Galphas-or Galphaq-coupled G-protein-coupled receptor on NMDA receptor selectively activates the NMDA receptor/Ca2+/calcineurin/cAMP response element-binding protein-regulated transcriptional coactivator 1 pathway to effectively induce brain-derived neurotrophic factor expression in neurons. J. Neurosci. 2015, 35, 5606–5624. [Google Scholar] [PubMed]
- De Chiara, V.; Angelucci, F.; Rossi, S.; Musella, A.; Cavasinni, F.; Cantarella, C.; Mataluni, G.; Sacchetti, L.; Napolitano, F.; Castelli, M.; et al. Brain-derived neurotrophic factor controls cannabinoid CB1 receptor function in the striatum. J. Neurosci. 2010, 30, 8127–8137. [Google Scholar] [CrossRef]
- Beaulieu, J.M.; Del’guidice, T.; Sotnikova, T.D.; Lemasson, M.; Gainetdinov, R.R. Beyond cAMP: The Regulation of Akt and GSK3 by Dopamine Receptors. Front. Mol. Neurosci. 2011, 4, 38. [Google Scholar] [CrossRef]
- Hur, E.M.; Zhou, F.Q. GSK3 signalling in neural development. Nat. Rev. Neurosci. 2010, 11, 539–551. [Google Scholar] [CrossRef]
- Matsuda, S.; Ikeda, Y.; Murakami, M.; Nakagawa, Y.; Tsuji, A.; Kitagishi, Y. Roles of PI3K/AKT/GSK3 Pathway Involved in Psychiatric Illnesses. Diseases 2019, 7, 22. [Google Scholar] [CrossRef]
- Park, H.; Poo, M.M. Neurotrophin regulation of neural circuit development and function. Nat. Rev. Neurosci. 2013, 14, 7–23. [Google Scholar] [CrossRef]
- Weickert, C.S.; Hyde, T.M.; Lipska, B.K.; Herman, M.M.; Weinberger, D.R.; Kleinman, J.E. Reduced brain-derived neurotrophic factor in prefrontal cortex of patients with schizophrenia. Mol. Psychiatry 2003, 8, 592–610. [Google Scholar] [CrossRef] [PubMed]
- Hashimoto, T.; Bergen, S.E.; Nguyen, Q.L.; Xu, B.; Monteggia, L.M.; Pierri, J.N.; Sun, Z.; Sampson, A.R.; Lewis, D.A. Relationship of brain-derived neurotrophic factor and its receptor TrkB to altered inhibitory prefrontal circuitry in schizophrenia. J. Neurosci. 2005, 25, 372–383. [Google Scholar] [CrossRef] [PubMed]
- Hong, E.J.; McCord, A.E.; Greenberg, M.E. A biological function for the neuronal activity-dependent component of Bdnf transcription in the development of cortical inhibition. Neuron 2008, 60, 610–624. [Google Scholar] [CrossRef] [PubMed]




© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Komatsu, H.; Fukuchi, M.; Habata, Y. Potential Utility of Biased GPCR Signaling for Treatment of Psychiatric Disorders. Int. J. Mol. Sci. 2019, 20, 3207. https://doi.org/10.3390/ijms20133207
Komatsu H, Fukuchi M, Habata Y. Potential Utility of Biased GPCR Signaling for Treatment of Psychiatric Disorders. International Journal of Molecular Sciences. 2019; 20(13):3207. https://doi.org/10.3390/ijms20133207
Chicago/Turabian StyleKomatsu, Hidetoshi, Mamoru Fukuchi, and Yugo Habata. 2019. "Potential Utility of Biased GPCR Signaling for Treatment of Psychiatric Disorders" International Journal of Molecular Sciences 20, no. 13: 3207. https://doi.org/10.3390/ijms20133207
APA StyleKomatsu, H., Fukuchi, M., & Habata, Y. (2019). Potential Utility of Biased GPCR Signaling for Treatment of Psychiatric Disorders. International Journal of Molecular Sciences, 20(13), 3207. https://doi.org/10.3390/ijms20133207

