Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

mTOR kinase activity disrupts a phosphorylation signaling network in schizophrenia brain

Abstract

The AKT-mTOR signaling transduction pathway plays an important role in neurodevelopment and synaptic plasticity. mTOR is a serine/threonine kinase that modulates signals from multiple neurotransmitters and phosphorylates specific proteins to regulate protein synthesis and cytoskeletal organization. There is substantial evidence demonstrating abnormalities in AKT expression and activity in different schizophrenia (SZ) models. However, direct evidence for dysregulated mTOR kinase activity and its consequences on downstream effector proteins in SZ pathophysiology is lacking. Recently, we reported reduced phosphorylation of mTOR at an activating site and abnormal mTOR complex formation in the SZ dorsolateral prefrontal cortex (DLPFC). Here, we expand on our hypothesis of disrupted mTOR signaling in the SZ brain and studied the expression and activity of downstream effector proteins of mTOR complexes and the kinase activity profiles of SZ subjects. We found that S6RP phosphorylation, downstream of mTOR complex I, is reduced, whereas PKCĪ± phosphorylation, downstream of mTOR complex II, is increased in SZ DLPFC. In rats chronically treated with haloperidol, we showed that S6RP phosphorylation is increased in the rat frontal cortex, suggesting a potential novel mechanism of action for antipsychotics. We also demonstrated key differences in kinase signaling networks between SZ and comparison subjects for both males and females using kinome peptide arrays. We further investigated the role of mTOR kinase activity by inhibiting it with rapamycin in postmortem tissue and compared the impact of mTOR inhibition in SZ and comparison subjects using kinome arrays. We found that SZ subjects are globally more sensitive to rapamycin treatment and AMP-activated protein kinase (AMPK) contributes to this differential kinase activity. Together, our findings provide new insights into the role of mTOR as a master regulator of kinase activity in SZ and suggest potential targets for therapeutic intervention.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Abnormal phosphorylation of downstream effector proteins of mTOR complexes in SZ DLPFC.
Fig. 2: Chronic antipsychotic treatment leads to increased phosphorylation of S6RP in rat brain.
Fig. 3: Rapamycin differentially inhibits the activity of mTOR and related kinases in SZ DLPFC.

Similar content being viewed by others

References

  1. Owen MJ, Sawa A, Mortensen PB. Schizophrenia. Lancet. 2016;388:86ā€“97.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  2. Funk AJ, McCullumsmith RE, Haroutunian V, Meador-Woodruff JH. Abnormal Activity of the MAPK- and cAMP-Associated Signaling Pathways in Frontal Cortical Areas in Postmortem Brain in Schizophrenia. Neuropsychopharmacology. 2012;37:896ā€“905.

    CASĀ  PubMedĀ  Google ScholarĀ 

  3. Kyosseva SV, Elbein AD, Griffin WST, Mrak RE, Lyon M, Karson CN. Mitogen-activated protein kinases in schizophrenia. Biol. Psychiatry. 1999;46:689ā€“96.

    CASĀ  PubMedĀ  Google ScholarĀ 

  4. Emamian ES. AKT/GSK3 signaling pathway and schizophrenia. Front. Mol. Neurosci. 2012;5:33.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  5. Emamian ES, Hall D, Birnbaum MJ, Karayiorgou M, Gogos JA. Convergent evidence for impaired AKT1-GSK3Ī² signaling in schizophrenia. Nat. Genet. 2004;36:131ā€“7.

    CASĀ  PubMedĀ  Google ScholarĀ 

  6. McGuire JL, Hammond JH, Yates SD, Chen D, Haroutunian V, Meador-Woodruff JH, et al. Altered serine/threonine kinase activity in schizophrenia. Brain Res. 2014;1568:42ā€“54.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  7. Chadha, R & Meador-Woodruff, JH Downregulated AKT-mTOR signaling pathway proteins in dorsolateral prefrontal cortex in Schizophrenia. Neuropsychopharmacology 1ā€“9 (2020) https://doi.org/10.1038/s41386-020-0614-2.

  8. Wang L, Zhou K, Fu Z, Yu D, Huang H, Zang X, et al. Brain Development and Akt Signaling: the Crossroads of Signaling Pathway and Neurodevelopmental Diseases. J. Mol. Neurosci. 2017;61:379ā€“84.

    CASĀ  PubMedĀ  Google ScholarĀ 

  9. Costa-Mattioli M, Monteggia LM. mTOR complexes in neurodevelopmental and neuropsychiatric disorders. Nat. Neurosci. 2013;16:1537ā€“43.

    CASĀ  PubMedĀ  Google ScholarĀ 

  10. McGuire JL, Depasquale EA, Funk AJ, Oā€™Donnovan SM, Hasselfeld K, Marwaha S, et al. Abnormalities of signal transduction networks in chronic schizophrenia. NPJ Schizophr. 2017;3:30.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  11. Bentea E, Depasquale EAKK, Oā€™donovan SM, Sullivan CR, Simmons M, Meador-Woodruff JH, et al. Kinase network dysregulation in a human induced pluripotent stem cell model of DISC1 schizophrenia. Mol. Omi. 2019;15:173ā€“88.

    CASĀ  Google ScholarĀ 

  12. Kozlovsky N, Shanon-Weickert C, Tomaskovic-Crook E, Kleinman JE, Belmaker RH, Agam G. Reduced GSK-3Ī² mRNA levels in postmortem dorsolateral prefrontal cortex of schizophrenic patients. J. Neural Transm. 2004;111:1583ā€“92.

    CASĀ  PubMedĀ  Google ScholarĀ 

  13. Gong R, Park CS, Abbassi NR, Tang S-J. Roles of glutamate receptors and the mammalian target of rapamycin (mTOR) signaling pathway in activity-dependent dendritic protein synthesis in hippocampal neurons. J. Biol. Chem. 2006;281:18802ā€“15.

    CASĀ  PubMedĀ  Google ScholarĀ 

  14. Hsu W-L, Chung H-W, Wu C-Y, Wu H-I, Lee Y-T, Chen E-C, et al. Glutamate Stimulates Local Protein Synthesis in the Axons of Rat Cortical Neurons by Activating Ī±-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) Receptors and Metabotropic Glutamate Receptors. J. Biol. Chem. 2015;290:20748ā€“60.

    CASĀ  PubMedĀ  Google ScholarĀ 

  15. English J, Fan Y, Fƶcking M, Lopez L, Hryniewiecka M, Wynne K, et al. Reduced protein synthesis in schizophrenia patient-derived olfactory cells. Transl. Psychiatry. 2015;5:e663.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  16. Huang W, Zhu PJ, Zhang S, Zhou H, Stoica L, Galiano M, et al. mTORC2 controls actin polymerization required for consolidation of long-term memory. Nat. Neurosci. 2013;16:441ā€“8.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  17. Ibarra-Lecue I, Diez-Alarcia R, Morentin B, Meana JJ, Callado LF, UrigĆ¼en L. Ribosomal Protein S6 Hypofunction in Postmortem Human Brain Links mTORC1-Dependent Signaling and Schizophrenia. Front. Pharmacol. 2020;11:344.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  18. Kim P, Scott MR, Meador-Woodruff JH. Abnormal expression of ER quality control and ER associated degradation proteins in the dorsolateral prefrontal cortex in schizophrenia. Schizophr. Res. 2018;197:484ā€“91.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  19. Kippe JM, Mueller TM, Haroutunian V, Meador-Woodruff JH. Abnormal N-acetylglucosaminyltransferase expression in prefrontal cortex in schizophrenia. Schizophr. Res. 2015;166:219ā€“24.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  20. Bauer DE, Haroutunian V, McCullumsmith RE, Meador-Woodruff JH. Expression of four housekeeping proteins in elderly patients with schizophrenia. J. Neural Transm. 2009;116:487ā€“91.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  21. Harte MK, Bachus SB, Reynolds GP. Increased N-acetylaspartate in rat striatum following long-term administration of haloperidol. Schizophr. Res. 2005;75:303ā€“8.

    CASĀ  PubMedĀ  Google ScholarĀ 

  22. Kashihara K, Sato M, Fujiwara Y, Harada T, Ogawa T, Otsuki S. Effects of intermittent and continuous haloperidol administration on the dopaminergic system in the rat brain. Biol. Psychiatry. 1986;21:650ā€“6.

    CASĀ  PubMedĀ  Google ScholarĀ 

  23. Bhambhvani HP, Mueller TM, Simmons MS, Meador-Woodruff JH. Actin polymerization is reduced in the anterior cingulate cortex of elderly patients with schizophrenia. Transl. Psychiatry. 2017;7:1278.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  24. Dorsett CR, McGuire JL, Niedzielko TL, Depasquale EAK, Meller J, Floyd CL, et al. Traumatic brain injury induces alterations in cortical glutamate uptake without a reduction in glutamate transporter-1 protein expression. J. Neurotrauma. 2017;34:220ā€“34.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  25. DePasquale EAK, Alganem K, Bentea E, Nawreen N, McGuire JL, Naji F, et al. KRSA: Network-based Prediction of Differential Kinase Activity from Kinome Array Data. bioRxiv. 2020. https://doi.org/10.1101/2020.08.26.268581. 2020.08.26.268581

    ArticleĀ  Google ScholarĀ 

  26. Xue Y, Liu Z, Gao X, Jin C, Wen L, Yao X, et al. GPS-SNO: Computational Prediction of Protein S-Nitrosylation Sites with a Modified GPS Algorithm. PLoS One. 2010;5:e11290.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  27. Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, et al. The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45:D362ā€“D368.

    CASĀ  PubMedĀ  Google ScholarĀ 

  28. Biever A, Valjent E, Puighermanal E. Ribosomal protein S6 phosphorylation in the nervous system: From regulation to function. Front. Mol. Neurosci. 2015;8:75.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  29. Hetman M, Slomnicki LP. Ribosomal biogenesis as an emerging target of neurodevelopmental pathologies. J. Neurochem. 2019;148:325ā€“47.

    CASĀ  PubMedĀ  Google ScholarĀ 

  30. Slomnicki LP, Pietrzak M, Vashishta A, Jones J, Lynch N, Elliot S, et al. Requirement of Neuronal Ribosome Synthesis for Growth and Maintenance of the Dendritic Tree. J. Biol. Chem. 2016;291:5721ā€“39.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  31. Bowling H, Zhang G, Bhattacharya A, PĆ©rez-Cuesta LM, Deinhardt K, Hoeffer CA, et al. Antipsychotics activate mTORC1-dependent translation to enhance neuronal morphological complexity. Sci. Signal. 2014;7:ra4.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  32. Valjent E, Bertran-Gonzalez J, Bowling H, Lopez S, Santini E, Matamales M, et al. Haloperidol regulates the state of phosphorylation of ribosomal protein S6 via activation of PKA and phosphorylation of DARPP-32. Neuropsychopharmacology. 2011;36:2561ā€“70.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  33. 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;72:197ā€“203.

    CASĀ  PubMedĀ  Google ScholarĀ 

  34. Pan B, Huang XF, Deng C. Aripiprazole and haloperidol activate GSK3Ī²-dependent signalling pathway differentially in various brain regions of rats. Int. J. Mol. Sci. 2016;17:459.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  35. Roh M-S, Seo MS, Kim Y, Kim SH, Jeon WJ, Ahn YM, et al. Haloperidol and clozapine differentially regulate signals upstream of glycogen synthase kinase 3 in the rat frontal cortex. Exp. Mol. Med. 2007;39:353ā€“60.

    CASĀ  PubMedĀ  Google ScholarĀ 

  36. Garza-LombĆ³ C, Schroder A, Reyes-Reyes EM, Franco R. mTOR/AMPK signaling in the brain: Cell metabolism, proteostasis and survival. Current Opinion in Toxicology. 2018;8:102ā€“10.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  37. Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, et al. AMPK Phosphorylation of Raptor Mediates a Metabolic Checkpoint. Mol. Cell. 2008;30:214ā€“26.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  38. Inoki K, Kim J, Guan K-L. AMPK and mTOR in Cellular Energy Homeostasis and Drug Targets. Annu. Rev. Pharmacol. Toxicol. 2012;52:381ā€“400.

    CASĀ  PubMedĀ  Google ScholarĀ 

  39. Rabanal-Ruiz Y, Otten EG, Korolchuk VI. MTORC1 as the main gateway to autophagy. Essays in Biochemistry. 2017;61:565ā€“84.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  40. Al Eissa MM, Fiorentino A, Sharp SI, Oā€™Brien NL, Wolfe K, Giaroli G, et al. Exome sequence analysis and follow up genotyping implicates rare ULK1 variants to be involved in susceptibility to schizophrenia. Ann. Hum. Genet. 2018;82:88ā€“92.

    CASĀ  PubMedĀ  Google ScholarĀ 

  41. Barnes MR, Huxley-Jones J, Maycox PR, Lennon M, Thornber A, Kelly F, et al. Transcription and pathway analysis of the superior temporal cortex and anterior prefrontal cortex in schizophrenia. J. Neurosci. Res. 2011;89:1218ā€“27.

    CASĀ  PubMedĀ  Google ScholarĀ 

  42. Kroemer G, MariƱo G, Levine B. Autophagy and the Integrated Stress Response. Molecular Cell. 2010;40:280ā€“93.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  43. Horesh Y, Katsel P, Haroutunian V, Domany E. Gene expression signature is shared by patients with Alzheimerā€™s disease and schizophrenia at the superior temporal gyrus. Eur. J. Neurol. 2011;18:410ā€“24.

    CASĀ  PubMedĀ  Google ScholarĀ 

  44. Glantz LA, Lewis DA. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch. Gen. Psychiatry. 2000;57:65ā€“73.

    CASĀ  PubMedĀ  Google ScholarĀ 

  45. Glausier JR, Lewis DA. Dendritic spine pathology in schizophrenia. Neuroscience. 2013;251:90ā€“107.

    CASĀ  PubMedĀ  Google ScholarĀ 

  46. Garey LJ, Ong WY, Patel TS, Kanani M, Davis A, Mortimer AM, et al. Reduced dendritic spine density on cerebral cortical pyramidal neurons in schizophrenia. J. Neurol. Neurosurg. Psychiatry. 1998;65:446ā€“53.

    CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  47. Lewis DA, Curley AA, Glausier JR, Volk DW. Cortical parvalbumin interneurons and cognitive dysfunction in schizophrenia. Trends in Neurosciences. 2012;35:57ā€“67.

    CASĀ  PubMedĀ  Google ScholarĀ 

  48. Rojas-BenĆ­tez D, Ibar C, Glavic Ɓ. The Drosophila EKC/KEOPS complex: Roles in protein synthesis homeostasis and animal growth. Fly (Austin). 2013;7:168ā€“72.

    Google ScholarĀ 

  49. Wu MF, Wang SG. Human TAO kinase 1 induces apoptosis in SH-SY5Y cells. Cell Biol. Int. 2008;32:151ā€“6.

    PubMedĀ  Google ScholarĀ 

  50. Pinner AL, Tucholski J, Haroutunian V, McCullumsmith RE, Meador-Woodruff JH. Decreased protein S-palmitoylation in dorsolateral prefrontal cortex in schizophrenia. Schizophr. Res. 2016;177:78ā€“87.

    PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  51. Mccullumsmith RE, Hammond JH, Shan D, Meador-Woodruff JH. Postmortem brain: An underutilized substrate for studying severe mental illness. Neuropsychopharmacology. 2014;39:65ā€“87.

    PubMedĀ  Google ScholarĀ 

Download references

Acknowledgements

The authors would like to thank Dr. Rosalinda Roberts and the Alabama Brain Collection for postmortem cortical samples used in assay development for immunoblotting analyses.

Funding

RC was supported by the Marie and Emmett Carmichael fund for graduate students in biosciences. KA and REM were supported by NIMH R01 MH107487 and MH121102.

Author information

Authors and Affiliations

Authors

Contributions

Authors RC and JMW designed the study. RC executed experimental protocols, performed data calculations, statistical analyses, and literature searches. RC performed the biochemical studies, whereas kinome array experiments were executed and analyzed by KA. RC wrote the first draft of the manuscript, followed by editing by JMW and REM. All authors contributed to and have approved the final manuscript.

Corresponding author

Correspondence to Radhika Chadha.

Ethics declarations

Conflict of interest

The authors declare no competing interests.

Additional information

Publisherā€™s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chadha, R., Alganem, K., Mccullumsmith, R.E. et al. mTOR kinase activity disrupts a phosphorylation signaling network in schizophrenia brain. Mol Psychiatry 26, 6868ā€“6879 (2021). https://doi.org/10.1038/s41380-021-01135-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-021-01135-9

This article is cited by

Search

Quick links