J Korean Neuropsychiatr Assoc.  2019 Feb;58(1):12-17. 10.4306/jknpa.2019.58.1.12.

New Insights into the Molecular and Cellular Mechanism of Antipsychotics

Affiliations
  • 1Department of Psychiatry, Seoul National University Hospital, Seoul, Korea. sh3491@snu.ac.kr

Abstract

The serendipitous discovery of drugs with antipsychotic action has provided a clue to understanding the pathogenesis of psychotic disorders and to develop so called "˜antipsychotics' drugs. From the "˜dopamine hypothesis' to current molecular psychiatry findings, researchers have endeavored to improve the understanding of the mechanism of action of antipsychotic drugs. Signaling molecules were examined as novel intracellular targets of antipsychotic action. The findings regarding the effects of antipsychotics on inflammation, metabolism, and cellular fate have helped conceptualize the biological action of antipsychotics. This review summarizes the current knowledge of the molecular psychiatry findings regarding the action mechanisms of antipsychotics. The accumulation of research findings will lead to a better understanding of the pathogenesis of schizophrenia and its treatment mechanism.

Keyword

Antipsychotics; Psychopharmacology; Signal transduction

MeSH Terms

Antipsychotic Agents*
Inflammation
Metabolism
Psychopharmacology
Psychotic Disorders
Schizophrenia
Signal Transduction
Antipsychotic Agents

Reference

1. Carlsson A, Lindqvist M. Effect of chlorpromazine or haloperidol on formation of 3methoxytyramine and normetanephrine in mouse brain. Acta Pharmacol Toxicol (Copenh). 1963; 20:140–144.
Article
2. Davis KL, Kahn RS, Ko G, Davidson M. Dopamine in schizophrenia: a review and reconceptualization. Am J Psychiatry. 1991; 148:1474–1486.
Article
3. Pickar D, Labarca R, Linnoila M, Roy A, Hommer D, Everett D, et al. Neuroleptic-induced decrease in plasma homovanillic acid and antipsychotic activity in schizophrenic patients. Science. 1984; 225:954–957.
Article
4. Creese I, Burt DR, Snyder SH. Dopamine receptor binding predicts clinical and pharmacological potencies of antischizophrenic drugs. J Neuropsychiatry Clin Neurosci. 1996; 8:223–226.
Article
5. Davis JM, Chen N, Glick ID. A meta-analysis of the efficacy of second-generation antipsychotics. Arch Gen Psychiatry. 2003; 60:553–564.
Article
6. Leucht S, Cipriani A, Spineli L, Mavridis D, Orey D, Richter F, et al. Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. Lancet. 2013; 382:951–962.
Article
7. Crilly J. The history of clozapine and its emergence in the US market: a review and analysis. Hist Psychiatry. 2007; 18:39–60.
Article
8. Hippius H. A historical perspective of clozapine. J Clin Psychiatry. 1999; 60:Suppl 12. 22–23.
9. Nucifora FC Jr, Mihaljevic M, Lee BJ, Sawa A. Clozapine as a model for antipsychotic development. Neurotherapeutics. 2017; 14:750–761.
Article
10. Amato D. Serotonin in antipsychotic drugs action. Behav Brain Res. 2015; 277:125–135.
Article
11. Meltzer HY. Serotonergic mechanisms as targets for existing and novel antipsychotics. Handb Exp Pharmacol. 2012; 87–124.
Article
12. Forray C, Buller R. Challenges and opportunities for the development of new antipsychotic drugs. Biochem Pharmacol. 2017; 143:10–24.
Article
13. Kondej M, Stepnicki P, Kaczor AA. Multi-target approach for drug discovery against schizophrenia. Int J Mol Sci. 2018; 19:E3105.
Article
14. Molteni R, Calabrese F, Racagni G, Fumagalli F, Riva MA. Antipsychotic drug actions on gene modulation and signaling mechanisms. Pharmacol Ther. 2009; 124:74–85.
Article
15. Bateup HS, Svenningsson P, Kuroiwa M, Gong S, Nishi A, Heintz N, et al. Cell type-specific regulation of DARPP-32 phosphorylation by psychostimulant and antipsychotic drugs. Nat Neurosci. 2008; 11:932–939.
Article
16. Fienberg AA, Hiroi N, Mermelstein PG, Song W, Snyder GL, Nishi A, et al. DARPP-32: regulator of the efficacy of dopaminergic neurotransmission. Science. 1998; 281:838–842.
Article
17. Wang H, Farhan M, Xu J, Lazarovici P, Zheng W. The involvement of DARPP-32 in the pathophysiology of schizophrenia. Oncotarget. 2017; 8:53791–53803.
Article
18. Masri B, Salahpour A, Didriksen M, Ghisi V, Beaulieu JM, Gainetdinov RR, et al. Antagonism of dopamine D2 receptor/beta-arrestin 2 interaction is a common property of clinically effective antipsychotics. Proc Natl Acad Sci U S A. 2008; 105:13656–13661.
Article
19. Urs NM, Gee SM, Pack TF, McCorvy JD, Evron T, Snyder JC, et al. Distinct cortical and striatal actions of a β-arrestin-biased dopamine D2 receptor ligand reveal unique antipsychotic-like properties. Proc Natl Acad Sci U S A. 2016; 113:E8178–E8186.
Article
20. Halene TB, Siegel SJ. Antipsychotic-like properties of phosphodiesterase 4 inhibitors: evaluation of 4-(3-butoxy-4-methoxybenzyl)-2-imidazolidinone (RO-20-1724) with auditory event-related potentials and prepulse inhibition of startle. J Pharmacol Exp Ther. 2008; 326:230–239.
Article
21. Lipina TV, Palomo V, Gil C, Martinez A, Roder JC. Dual inhibitor of PDE7 and GSK-3-VP1.15 acts as antipsychotic and cognitive enhancer in C57BL/6J mice. Neuropharmacology. 2013; 64:205–214.
Article
22. Pandey MK, DeGrado TR. Glycogen synthase kinase-3 (GSK-3)-targeted therapy and imaging. Theranostics. 2016; 6:571–593.
Article
23. Wiescholleck V, Manahan-Vaughan D. PDE4 inhibition enhances hippocampal synaptic plasticity in vivo and rescues MK801-induced impairment of long-term potentiation and object recognition memory in an animal model of psychosis. Transl Psychiatry. 2012; 2:e89.
Article
24. Hanson ND, Owens MJ, Nemeroff CB. Depression, antidepressants, and neurogenesis: a critical reappraisal. Neuropsychopharmacology. 2011; 36:2589–2602.
Article
25. Kusumi I, Boku S, Takahashi Y. Psychopharmacology of atypical antipsychotic drugs: from the receptor binding profile to neuroprotection and neurogenesis. Psychiatry Clin Neurosci. 2015; 69:243–258.
Article
26. Nandra KS, Agius M. The differences between typical and atypical antipsychotics: the effects on neurogenesis. Psychiatr Danub. 2012; 24:Suppl 1. S95–S99.
27. Gassó P, Mas S, Molina O, Bernardo M, Lafuente A, Parellada E. Neurotoxic/neuroprotective activity of haloperidol, risperidone and paliperidone in neuroblastoma cells. Prog Neuropsychopharmacol Biol Psychiatry. 2012; 36:71–77.
Article
28. Dakir EH, Pickard A, Srivastava K, McCrudden CM, Gross SR, Lloyd S, et al. The anti-psychotic drug pimozide is a novel chemotherapeutic for breast cancer. Oncotarget. 2018; 9:34889–34910.
Article
29. Roney MSI, Park SK. Antipsychotic dopamine receptor antagonists, cancer, and cancer stem cells. Arch Pharm Res. 2018; 41:384–408.
Article
30. Wu CH, Bai LY, Tsai MH, Chu PC, Chiu CF, Chen MY, et al. Pharmacological exploitation of the phenothiazine antipsychotics to develop novel antitumor agents-a drug repurposing strategy. Sci Rep. 2016; 6:27540.
Article
31. Zhang W, Zhang C, Liu F, Mao Y, Xu W, Fan T, et al. Antiproliferative activities of the second-generation antipsychotic drug sertindole against breast cancers with a potential application for treatment of breast-to-brain metastases. Sci Rep. 2018; 8:15753.
Article
32. Sachlos E, Risueño RM, Laronde S, Shapovalova Z, Lee JH, Russell J, et al. Identification of drugs including a dopamine receptor antagonist that selectively target cancer stem cells. Cell. 2012; 149:1284–1297.
Article
33. Frade JM, Ovejero-Benito MC. Neuronal cell cycle: the neuron itself and its circumstances. Cell Cycle. 2015; 14:712–720.
Article
34. Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med. 2013; 19:983–997.
Article
35. Merenlender-Wagner A, Malishkevich A, Shemer Z, Udawela M, Gibbons A, Scarr E, et al. Autophagy has a key role in the pathophysiology of schizophrenia. Mol Psychiatry. 2015; 20:126–132.
Article
36. Merenlender-Wagner A, Shemer Z, Touloumi O, Lagoudaki R, Giladi E, Andrieux A, et al. New horizons in schizophrenia treatment: autophagy protection is coupled with behavioral improvements in a mouse model of schizophrenia. Autophagy. 2014; 10:2324–2332.
Article
37. Schneider JL, Miller AM, Woesner ME. Autophagy and schizophrenia: a closer look at how dysregulation of neuronal cell homeostasis influences the pathogenesis of schizophrenia. Einstein J Biol Med. 2016; 31:34–39.
Article
38. Ranjan A, Srivastava SK. Penfluridol suppresses pancreatic tumor growth by autophagy-mediated apoptosis. Sci Rep. 2016; 6:26165.
Article
39. Shin SY, Lee KS, Choi YK, Lim HJ, Lee HG, Lim Y, et al. The antipsychotic agent chlorpromazine induces autophagic cell death by inhibiting the Akt/mTOR pathway in human U-87MG glioma cells. Carcinogenesis. 2013; 34:2080–2089.
Article
40. Yin YC, Lin CC, Chen TT, Chen JY, Tsai HJ, Wang CY, et al. Clozapine induces autophagic cell death in non-small cell lung cancer cells. Cell Physiol Biochem. 2015; 35:945–956.
Article
41. Kim SH, Park S, Yu HS, Ko KH, Park HG, Kim YS. The antipsychotic agent clozapine induces autophagy via the AMPK-ULK1-Beclin1 signaling pathway in the rat frontal cortex. Prog Neuropsychopharmacol Biol Psychiatry. 2018; 81:96–104.
Article
42. Choi Y, Jeong HJ, Liu QF, Oh ST, Koo BS, Kim Y, et al. Clozapine improves memory impairment and reduces Aβ level in the Tg-APPswe/PS1dE9 mouse model of Alzheimer’s disease. Mol Neurobiol. 2017; 54:450–460.
Article
43. Tourjman V, Kouassi É, Koué MÈ, Rocchetti M, Fortin-Fournier S, Fusar-Poli P, et al. Antipsychotics’ effects on blood levels of cytokines in schizophrenia: a meta-analysis. Schizophr Res. 2013; 151:43–47.
Article
44. Müller N, Myint AM, Krause D, Weidinger E, Schwarz MJ. Anti-inflammatory treatment in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2013; 42:146–153.
Article
45. Jeon S, Kim SH, Shin SY, Lee YH. Clozapine reduces Toll-like receptor 4/NF-κB-mediated inflammatory responses through inhibition of calcium/calmodulin-dependent Akt activation in microglia. Prog Neuropsychopharmacol Biol Psychiatry. 2018; 81:477–487.
Article
46. Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the link between insulin resistance, obesity and diabetes. Trends Immunol. 2004; 25:4–7.
Article
47. Fonseka TM, Müller DJ, Kennedy SH. Inflammatory cytokines and antipsychotic-induced weight gain: review and clinical implications. Mol Neuropsychiatry. 2016; 2:1–14.
Article
48. Cui D, Peng Y, Zhang C, Li Z, Su Y, Qi Y, et al. Macrophage migration inhibitory factor mediates metabolic dysfunction induced by atypical antipsychotic therapy. J Clin Invest. 2018; 128:4997–5007.
Article
49. Kim SF, Huang AS, Snowman AM, Teuscher C, Snyder SH. From the cover: antipsychotic drug-induced weight gain mediated by histamine H1 receptor-linked activation of hypothalamic AMP-kinase. Proc Natl Acad Sci U S A. 2007; 104:3456–3459.
Article
50. Kim MK, Kim SH, Yu HS, Park HG, Kang UG, Ahn YM, et al. The effect of clozapine on the AMPK-ACC-CPT1 pathway in the rat frontal cortex. Int J Neuropsychopharmacol. 2012; 15:907–917.
Article
51. Girgis RR, Javitch JA, Lieberman JA. Antipsychotic drug mechanisms: links between therapeutic effects, metabolic side effects and the insulin signaling pathway. Mol Psychiatry. 2008; 13:918–929.
Article
52. Guidotti A, Dong E, Kundakovic M, Satta R, Grayson DR, Costa E. Characterization of the action of antipsychotic subtypes on valproate-induced chromatin remodeling. Trends Pharmacol Sci. 2009; 30:55–60.
Article
53. Hyman SE. Target practice: HDAC inhibitors for schizophrenia. Nat Neurosci. 2012; 15:1180–1181.
Article
54. Kurita M, Holloway T, García-Bea A, Kozlenkov A, Friedman AK, Moreno JL, et al. HDAC2 regulates atypical antipsychotic responses through the modulation of mGlu2 promoter activity. Nat Neurosci. 2012; 15:1245–1254.
Article
55. De la Fuente Revenga M, Ibi D, Saunders JM, Cuddy T, Ijaz MK, Toneatti R, et al. HDAC2-dependent antipsychotic-like effects of chronic treatment with the HDAC Inhibitor SAHA in mice. Neuroscience. 2018; 388:102–117.
Article
56. Ibi D, De la Fuente Revenga M, Kezunovic N, Muguruza C, Saunders JM, Gaitonde SA, et al. Antipsychotic-induced Hdac2 transcription via NF-κB leads to synaptic and cognitive side effects. Nat Neurosci. 2017; 20:1247–1259.
Article
57. Swathy B, Banerjee M. Understanding epigenetics of schizophrenia in the backdrop of its antipsychotic drug therapy. Epigenomics. 2017; 9:721–736.
Article
58. Dong E, Grayson DR, Guidotti A, Costa E. Antipsychotic subtypes can be characterized by differences in their ability to modify GABAergic promoter methylation. Epigenomics. 2009; 1:201–211.
Article
59. Dong E, Nelson M, Grayson DR, Costa E, Guidotti A. Clozapine and sulpiride but not haloperidol or olanzapine activate brain DNA demethylation. Proc Natl Acad Sci U S A. 2008; 105:13614–13619.
Article
60. Guidotti A, Grayson DR. DNA methylation and demethylation as targets for antipsychotic therapy. Dialogues Clin Neurosci. 2014; 16:419–429.
Article
61. Kim SH, Lee HY, Yi H, Ahn YM, Kim YS. Haloperidol induces demethylation and expression of the dual specificity phosphatase 6 gene in MIA PaCa-2 human pancreatic cancer cells. Life Sci. 2012; 91:1317–1322.
Article
62. Luoni A, Riva MA. MicroRNAs and psychiatric disorders: from aetiology to treatment. Pharmacol Ther. 2016; 167:13–27.
Article
63. Swathy B, Saradalekshmi KR, Nair IV, Nair C, Banerjee M. Pharmacoepigenomic responses of antipsychotic drugs on pharmacogenes are likely to be modulated by miRNAs. Epigenomics. 2017; 9:811–821.
Article
Full Text Links
  • JKNA
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