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.

  • Letter
  • Published:

Decreased expression of synapse-related genes and loss of synapses in major depressive disorder

Abstract

Previous imaging and postmortem studies have reported a lower brain volume and a smaller size and density of neurons in the dorsolateral prefrontal cortex (dlPFC) of subjects with major depressive disorder (MDD)1,2. These findings suggest that synapse number and function are decreased in the dlPFC of patients with MDD. However, there has been no direct evidence reported for synapse loss in MDD, and the gene expression alterations underlying these effects have not been identified. Here we use microarray gene profiling and electron microscopic stereology to reveal lower expression of synaptic-function–related genes (CALM2, SYN1, RAB3A, RAB4B and TUBB4) in the dlPFC of subjects with MDD and a corresponding lower number of synapses. We also identify a transcriptional repressor, GATA1, expression of which is higher in MDD and that, when expressed in PFC neurons, is sufficient to decrease the expression of synapse-related genes, cause loss of dendritic spines and dendrites, and produce depressive behavior in rat models of depression.

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

Figure 1: Synaptic-function–related genes, the number of spine synapses and MAP2 expression are decreased in the dlPFC of subjects with MDD.
Figure 2: Identification of GATA1 as a transcriptional repressor of synapse-related genes.
Figure 3: GATA1 overexpression in cortical neurons decreases Rab4b expression and reduces dendrite branching.
Figure 4: Viral expression of GATA1 in rat PFCs causes depressive behavior.

Similar content being viewed by others

References

  1. Rajkowska, G. et al. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol. Psychiatry 45, 1085–1098 (1999).

    Article  CAS  Google Scholar 

  2. Drevets, W.C. Functional anatomical abnormalities in limbic and prefrontal cortical structures in major depression. Prog. Brain Res. 126, 413–431 (2000).

    Article  CAS  Google Scholar 

  3. Lecrubier, Y. The burden of depression and anxiety in general medicine. J. Clin. Psychiatry 62 (suppl. 8), 4–9 (2001).

    Google Scholar 

  4. Boyle, L.L., Porsteinsson, A.P., Cui, X., King, D.A. & Lyness, J.M. Depression predicts cognitive disorders in older primary care patients. J. Clin. Psychiatry 71, 74–79 (2010).

    Article  Google Scholar 

  5. Barabassy, A., Beinhoff, U. & Riepe, M.W. Cognitive estimation in aged patients with major depressive disorder. Psychiatry Res. 176, 26–29 (2010).

    Article  Google Scholar 

  6. Levens, S.M. & Gotlib, I.H. Impaired selection of relevant positive information in depression. Depress. Anxiety 26, 403–410 (2009).

    Article  Google Scholar 

  7. Radley, J.J. et al. Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex. Cereb. Cortex 16, 313–320 (2006).

    Article  Google Scholar 

  8. Liu, R.J. & Aghajanian, G.K. Stress blunts serotonin- and hypocretin-evoked EPSCs in prefrontal cortex: role of corticosterone-mediated apical dendritic atrophy. Proc. Natl. Acad. Sci. USA 105, 359–364 (2008).

    Article  CAS  Google Scholar 

  9. Bianchi, M., Hagan, J.J. & Heidbreder, C.A. Neuronal plasticity, stress and depression: involvement of the cytoskeletal microtubular system? Curr. Drug Targets CNS Neurol. Disord. 4, 597–611 (2005).

    Article  CAS  Google Scholar 

  10. Drevets, W.C. et al. Subgenual prefrontal cortex abnormalities in mood disorders. Nature 386, 824–827 (1997).

    Article  CAS  Google Scholar 

  11. Kang, H.J. et al. Gene expression profiling in postmortem prefrontal cortex of major depressive disorder. J. Neurosci. 27, 13329–13340 (2007).

    Article  CAS  Google Scholar 

  12. DeLorenzo, R.J. & Freedman, S.D. Calcium dependent neurotransmitter release and protein phosphorylation in synaptic vesicles. Biochem. Biophys. Res. Commun. 80, 183–192 (1978).

    Article  CAS  Google Scholar 

  13. De Camilli, P. & Greengard, P. Synapsin I: a synaptic vesicle-associated neuronal phosphoprotein. Biochem. Pharmacol. 35, 4349–4357 (1986).

    Article  CAS  Google Scholar 

  14. Elferink, L.A. & Scheller, R.H. Synaptic vesicle proteins and regulated exocytosis. Prog. Brain Res. 105, 79–85 (1995).

    Article  CAS  Google Scholar 

  15. Wigge, P. & McMahon, H.T. The amphiphysin family of proteins and their role in endocytosis at the synapse. Trends Neurosci. 21, 339–344 (1998).

    Article  CAS  Google Scholar 

  16. Belizaire, R. et al. Characterization of synaptogyrin 3 as a new synaptic vesicle protein. J. Comp. Neurol. 470, 266–281 (2004).

    Article  CAS  Google Scholar 

  17. Zhong, L., Cherry, T., Bies, C.E., Florence, M.A. & Gerges, N.Z. Neurogranin enhances synaptic strength through its interaction with calmodulin. EMBO J. 28, 3027–3039 (2009).

    Article  CAS  Google Scholar 

  18. Brown, T.C., Correia, S.S., Petrok, C.N. & Esteban, J.A. Functional compartmentalization of endosomal trafficking for the synaptic delivery of AMPA receptors during long-term potentiation. J. Neurosci. 27, 13311–13315 (2007).

    Article  CAS  Google Scholar 

  19. Hoffman, P.N. Distinct roles of neurofilament and tubulin gene expression in axonal growth. Ciba Found. Symp. 138, 192–204 (1988).

    CAS  Google Scholar 

  20. Kim, S. & Webster, M.J. Correlation analysis between genome-wide expression profiles and cytoarchitectural abnormalities in the prefrontal cortex of psychiatric disorders. Mol. Psychiatry 15, 326–336 (2010).

    Article  CAS  Google Scholar 

  21. Kim, S. & Webster, M.J. Integrative genome-wide association analysis of cytoarchitectural abnormalities in the prefrontal cortex of psychiatric disorders. Mol. Psychiatry 16, 452–461 (2011).

    Article  CAS  Google Scholar 

  22. Tochigi, M. et al. Gene expression profiling of major depression and suicide in the prefrontal cortex of postmortem brains. Neurosci. Res. 60, 184–191 (2008).

    Article  CAS  Google Scholar 

  23. Sequeira, A. et al. Global brain gene expression analysis links glutamatergic and GABAergic alterations to suicide and major depression. PLoS ONE 4, e6585 (2009).

    Article  Google Scholar 

  24. Aston, C., Jiang, L. & Sokolov, B.P. Transcriptional profiling reveals evidence for signaling and oligodendroglial abnormalities in the temporal cortex from patients with major depressive disorder. Mol. Psychiatry 10, 309–322 (2005).

    Article  CAS  Google Scholar 

  25. Tsang, A.P. et al. FOG, a multitype zinc finger protein, acts as a cofactor for transcription factor GATA-1 in erythroid and megakaryocytic differentiation. Cell 90, 109–119 (1997).

    Article  CAS  Google Scholar 

  26. Johnson, K.D., Kim, S.I. & Bresnick, E.H. Differential sensitivities of transcription factor target genes underlie cell type-specific gene expression profiles. Proc. Natl. Acad. Sci. USA 103, 15939–15944 (2006).

    Article  CAS  Google Scholar 

  27. Willner, P. Validity, reliability and utility of the chronic mild stress model of depression: a 10-year review and evaluation. Psychopharmacology (Berl.) 134, 319–329 (1997).

    Article  CAS  Google Scholar 

  28. Baldelli, P., Fassio, A., Valtorta, F. & Benfenati, F. Lack of synapsin I reduces the readily releasable pool of synaptic vesicles at central inhibitory synapses. J. Neurosci. 27, 13520–13531 (2007).

    Article  CAS  Google Scholar 

  29. Geppert, M., Goda, Y., Stevens, C.F. & Sudhof, T.C. The small GTP-binding protein Rab3A regulates a late step in synaptic vesicle fusion. Nature 387, 810–814 (1997).

    Article  CAS  Google Scholar 

  30. Matthews, G. Calcium/calmodulin: a synaptic antidepressant? Neuron 32, 962–963 (2001).

    Article  CAS  Google Scholar 

  31. Pang, Z.P., Cao, P., Xu, W. & Sudhof, T.C. Calmodulin controls synaptic strength via presynaptic activation of calmodulin kinase II. J. Neurosci. 30, 4132–4142 (2010).

    Article  CAS  Google Scholar 

  32. Hoogenraad, C.C. et al. Neuron specific Rab4 effector GRASP-1 coordinates membrane specialization and maturation of recycling endosomes. PLoS Biol. 8, e1000283 (2010).

    Article  Google Scholar 

  33. Gorwood, P. [Severe depression: genes and the environment]. Encephale 35 (suppl. 7), S306–S309 (2009).

    Article  Google Scholar 

  34. Hammen, C. Stress and depression. Annu. Rev. Clin. Psychol. 1, 293–319 (2005).

    Article  Google Scholar 

  35. Viger, R.S., Guittot, S.M., Anttonen, M., Wilson, D.B. & Heikinheimo, M. Role of the GATA family of transcription factors in endocrine development, function, and disease. Mol. Endocrinol. 22, 781–798 (2008).

    Article  CAS  Google Scholar 

  36. Nardelli, J., Thiesson, D., Fujiwara, Y., Tsai, F.Y. & Orkin, S.H. Expression and genetic interaction of transcription factors GATA-2 and GATA-3 during development of the mouse central nervous system. Dev. Biol. 210, 305–321 (1999).

    Article  CAS  Google Scholar 

  37. Kala, K. et al. Gata2 is a tissue-specific post-mitotic selector gene for midbrain GABAergic neurons. Development 136, 253–262 (2009).

    Article  CAS  Google Scholar 

  38. El Wakil, A., Francius, C., Wolff, A., Pleau-Varet, J. & Nardelli, J. The GATA2 transcription factor negatively regulates the proliferation of neuronal progenitors. Development 133, 2155–2165 (2006).

    Article  CAS  Google Scholar 

  39. Cole, S.W. et al. Computational identification of gene-social environment interaction at the human IL6 locus. Proc. Natl. Acad. Sci. USA 107, 5681–5686 (2010).

    Article  CAS  Google Scholar 

  40. Li, N. et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 329, 959–964 (2010).

    Article  CAS  Google Scholar 

  41. Hajszan, T. et al. Remodeling of hippocampal spine synapses in the rat learned helplessness model of depression. Biol. Psychiatry 65, 392–400 (2009).

    Article  Google Scholar 

  42. Hajszan, T. et al. Effects of estradiol on learned helplessness and associated remodeling of hippocampal spine synapses in female rats. Biol. Psychiatry 67, 168–174 (2010).

    Article  CAS  Google Scholar 

  43. Noh, J.S. & Gwag, B.J. Attenuation of oxidative neuronal necrosis by a dopamine D1 agonist in mouse cortical cell cultures. Exp. Neurol. 146, 604–608 (1997).

    Article  CAS  Google Scholar 

  44. Sholl, D.A. Dendritic organization in the neurons of the visual and motor cortices of the cat. J. Anat. 87, 387–406 (1953).

    CAS  Google Scholar 

  45. Valentine, G., Dow, A., Banasr, M., Pittman, B. & Duman, R. Differential effects of chronic antidepressant treatment on shuttle box escape deficits induced by uncontrollable stress. Psychopharmacology (Berl.) 200, 585–596 (2008).

    Article  CAS  Google Scholar 

  46. Banasr, M. et al. Chronic unpredictable stress decreases cell proliferation in the cerebral cortex of the adult rat. Biol. Psychiatry 62, 496–504 (2007).

    Article  CAS  Google Scholar 

  47. Pliakas, A.M. et al. Altered responsiveness to cocaine and increased immobility in the forced swim test associated with elevated cAMP response element-binding protein expression in nucleus accumbens. J. Neurosci. 21, 7397–7403 (2001).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work is supported by US Public Health Service grants MH45481 (R.S.D.), 2 P01 MH25642 (R.S.D.), MH67996 (C.A.S.) and P20 RR17701 (C.A.S.), the Connecticut Mental Health Center (R.S.D.) and a National Research Foundation of Korea grant (NRF, 2011-0028317; H.S.). We acknowledge the invaluable contributions made by the families consenting to donate brain tissue and be interviewed. We also thank the Cuyahoga County Coroner and staff, Cleveland, Ohio, for their willing assistance. We thank J. Overholser, G. Jurjus, H. Meltzer, L. Konick, L. Dieter, N. Herbst, G. Mahajan, H. Kooiman and J. Cobb for their contributions to the psychiatric assessment and human tissue dissection and preparation. We thank T.H. Kim (Yale University) for his advice and suggestions regarding the binding motif analysis and chromatin immunoprecipitation. The pXM-GATA1 plasmid was provided by A.B. Cantor (Harvard University), and the pAAV-eGFP-pA vector was provided by R.J. Dileone (Yale University). The GATA1-specific antibody was provided by E.H. Bresnick (University of Wisconsin).

Author information

Authors and Affiliations

Authors

Contributions

H.J.K. was involved in planning and conducting all aspects of the research, including the analysis of microarray data, the confirmation of results, the molecular and cellular experiments and the construction of the viral vectors; H.J.K. also prepared the first draft of the manuscript. B.V. designed viral vectors, conducted the behavioral studies and was involved in the analysis and interpretation of rodent behavioral studies. C.A.S. and G.R. were responsible for the generation of tables containing the relevant information from the human subjects and the corresponding methodology and for preparation of human tissue and microdissections. T.H. conducted electron microscopy analysis of synapse number in postmortem tissue. M.B., with the technical help of A.L., carried out the rat CUS studies and prepared tissues for in situ hybridization analysis, surgeries for viral infusion in rats and supervised behavioral studies. P.L. was involved in the viral vector preparations. H.S. was involved in viral vector experiments, including surgical infusions, behavioral studies and immunohistochemistry. M.S.M. and L.S.J. synthesized the GATA1-specific inhibitor, K-7174. R.S.D. was responsible for overseeing the study, including all aspects of study design, data analysis, interpretation of results and preparation of the manuscript and figures. All authors discussed the results presented in the manuscript.

Corresponding author

Correspondence to Ronald S Duman.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 515 kb)

Supplementary Table 1

Synaptic enriched genes were decreased in MDD (XLS 28 kb)

Supplementary Table 2

TRANSFAC MATRIX of the synapse related genes_Human (XLS 6368 kb)

Supplementary Table 3

TRNASFAC MATRIX of the synapse related genes_Rat (XLS 4874 kb)

Supplementary Table 4

List of PCR primers used in this study for validation (XLS 33 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kang, H., Voleti, B., Hajszan, T. et al. Decreased expression of synapse-related genes and loss of synapses in major depressive disorder. Nat Med 18, 1413–1417 (2012). https://doi.org/10.1038/nm.2886

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2886

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing