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.

  • Resource
  • Published:

Single-nucleus transcriptomics of the prefrontal cortex in major depressive disorder implicates oligodendrocyte precursor cells and excitatory neurons

Abstract

Major depressive disorder (MDD) has an enormous impact on global disease burden, affecting millions of people worldwide and ranking as a leading cause of disability for almost three decades. Past molecular studies of MDD employed bulk homogenates of postmortem brain tissue, which obscures gene expression changes within individual cell types. Here we used single-nucleus transcriptomics to examine ~80,000 nuclei from the dorsolateral prefrontal cortex of male individuals with MDD (n = 17) and of healthy controls (n = 17). We identified 26 cellular clusters, and over 60% of these showed differential gene expression between groups. We found that the greatest dysregulation occurred in deep layer excitatory neurons and immature oligodendrocyte precursor cells (OPCs), and these contributed almost half (47%) of all changes in gene expression. These results highlight the importance of dissecting cell-type-specific contributions to the disease and offer opportunities to identify new avenues of research and novel targets for treatment.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Experimental flow.
Fig. 2: Identification of cell types.
Fig. 3: DEGs.
Fig. 4: Differential expression and biological associations.
Fig. 5: WGCNA.
Fig. 6: Contributions of OPC2 and Ex7.

Similar content being viewed by others

Data availability

Raw sequencing data, annotated gene–barcode matrix and lists of cells used for differential gene expression analysis are accessible on GEO using the accession number GSE144136. RNAScope and high-throughput qPCR data are available upon request.

Code availability

A sample custom R script (Supplementary_R_Script_1.R) used for analyzing high-throughput qPCR data is provided and an R script used to test the statistical significance of CCInx interactions is provided (Supplementary_R_Script_2.R) along with this paper.

References

  1. Depression and Other Common Mental Disorders: Global Health Estimates (World Health Organization, 2017).

  2. Wray, N. R. et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat. Genet. 50, 668–681 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Jansen, R. et al. Gene expression in major depressive disorder. Mol. Psychiatry 21, 339–347 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. 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  PubMed  PubMed Central  CAS  Google Scholar 

  5. Abdallah, C. G., Sanacora, G., Duman, R. S. & Krystal, J. H. The neurobiology of depression, ketamine and rapid-acting antidepressants: is it glutamate inhibition or activation? Pharmacol. Ther. 190, 148–158 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pantazatos, S. P. et al. Whole-transcriptome brain expression and exon-usage profiling in major depression and suicide: evidence for altered glial, endothelial and ATPase activity. Mol. Psychiatry 22, 760–773 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Edgar, N. & Sibille, E. A putative functional role for oligodendrocytes in mood regulation. Transl Psychiatry 2, e109 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Nagy, C. et al. Astrocytic abnormalities and global DNA methylation patterns in depression and suicide. Mol. Psychiatry 20, 320–328 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Duman, R. S., Aghajanian, G. K., Sanacora, G. & Krystal, J. H. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nat. Med. 22, 238–249 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lake, B. B. et al. Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain. Nat. Biotechnol. 36, 70–80 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mancarci, B. O. et al. Cross-laboratory analysis of brain cell type transcriptomes with applications to interpretation of bulk tissue data. eNeuro 4, ENEURO.0212-17.2017 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lake, B. B. et al. A comparative strategy for single-nucleus and single-cell transcriptomes confirms accuracy in predicted cell-type expression from nuclear RNA. Sci. Rep. 7, 6031 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Renthal, W. et al. Characterization of human mosaic Rett syndrome brain tissue by single-nucleus RNA sequencing. Nat. Neurosci. 21, 1670–1679 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mathys, H. et al. Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 570, 332–337 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Velmeshev, D. et al. Single-cell genomics identifies cell type-specific molecular changes in autism. Science 364, 685–689 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Northoff, G. & Sibille, E. Why are cortical GABA neurons relevant to internal focus in depression? A cross-level model linking cellular, biochemical and neural network findings. Mol. Psychiatry 19, 966–977 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sofroniew, M. & Vinters, H. Astrocytes: biology and pathology. Acta Neuropathol. 119, 7–35 (2010).

    Article  PubMed  Google Scholar 

  21. Anderson, M. A., Ao, Y. & Sofroniew, M. V. Heterogeneity of reactive astrocytes. Neurosci. Lett. 565, 23–29 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Thomas, P. D. et al. Applications for protein sequence-function evolution data: mRNA/protein expression analysis and coding SNP scoring tools. Nucleic Acids Res. 34, W645–W650 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Butts, B. D., Houde, C. & Mehmet, H. Maturation-dependent sensitivity of oligodendrocyte lineage cells to apoptosis: implications for normal development and disease. Cell Death Differ. 15, 1178–1186 (2008).

    Article  CAS  PubMed  Google Scholar 

  25. Jakel, S. et al. Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature 566, 543–547 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li, Q. S., Tian, C., Seabrook, G. R., Drevets, W. C. & Narayan, V. A. Analysis of 23andMe antidepressant efficacy survey data: implication of circadian rhythm and neuroplasticity in bupropion response. Transl Psychiatry 6, e889 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gutierrez-Sacristan, A. et al. PsyGeNET: a knowledge platform on psychiatric disorders and their genes. Bioinformatics 31, 3075–3077 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pinero, J. et al. DisGeNET: a comprehensive platform integrating information on human disease-associated genes and variants. Nucleic Acids Res. 45, D833–D839 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Szklarczyk, D. et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D607–D613 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Wochnik, G. M. et al. FK506-binding proteins 51 and 52 differentially regulate dynein interaction and nuclear translocation of the glucocorticoid receptor in mammalian cells. J. Biol. Chem. 280, 4609–4616 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Birey, F. et al. Genetic and stress-induced loss of NG2 glia triggers emergence of depressive-like behaviors through reduced secretion of FGF2. Neuron 88, 941–956 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mason, J. L. & Goldman, J. E. A2B5+ and O4+ cycling progenitors in the adult forebrain white matter respond differentially to PDGF-AA, FGF-2, and IGF-1. Mol. Cell. Neurosci. 20, 30–42 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Shulha, H. P. et al. Human-specific histone methylation signatures at transcription start sites in prefrontal neurons. PLoS Biol. 10, e1001427 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Spitzer, S. O. et al. Oligodendrocyte progenitor cells become regionally diverse and heterogeneous with age. Neuron 101, 459–471.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Psachoulia, K., Jamen, F., Young, K. M. & Richardson, W. D. Cell cycle dynamics of NG2 cells in the postnatal and ageing brain. Neuron Glia Biol. 5, 57–67 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Bergles, D. E., Jabs, R. & Steinhauser, C. Neuron–glia synapses in the brain. Brain Res. Rev. 63, 130–137 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Birey, F., Kokkosis, A. G. & Aguirre, A. Oligodendroglia-lineage cells in brain plasticity, homeostasis and psychiatric disorders. Curr. Opin. Neurobiol. 47, 93–103 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ge, W. P. et al. Long-term potentiation of neuron–glia synapses mediated by Ca2+-permeable AMPA receptors. Science 312, 1533–1537 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Ueno, H., Huang, X., Tanaka, Y. & Hirokawa, N. KIF16B/Rab14 molecular motor complex is critical for early embryonic development by transporting FGF receptor. Dev. Cell 20, 60–71 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Turner, C. A., Watson, S. J. & Akil, H. The fibroblast growth factor family: neuromodulation of affective behavior. Neuron 76, 160–174 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Turecki, G. & Meaney, M. J. Effects of the social environment and stress on glucocorticoid receptor gene methylation: a systematic review. Biol. Psychiatry 79, 87–96 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Zuehlke, A. D., Beebe, K., Neckers, L. & Prince, T. Regulation and function of the human HSP90AA1 gene. Gene 570, 8–16 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Huang, J. Y., Lynn Miskus, M. & Lu, H. C. FGF–FGFR mediates the activity-dependent dendritogenesis of layer IV neurons during barrel formation. J. Neurosci. 37, 12094–12105 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Pittenger, C. & Duman, R. S. Stress, depression, and neuroplasticity: a convergence of mechanisms. Neuropsychopharmacology 33, 88–109 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Caiati, M. D. et al. PrPC controls via protein kinase A the direction of synaptic plasticity in the immature hippocampus. J. Neurosci. 33, 2973–2983 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sevilla, L. M., Nachat, R., Groot, K. R. & Watt, F. M. Kazrin regulates keratinocyte cytoskeletal networks, intercellular junctions and differentiation. J. Cell Sci. 121, 3561–3569 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Bribian, A. et al. Role of the cellular prion protein in oligodendrocyte precursor cell proliferation and differentiation in the developing and adult mouse CNS. PLoS ONE 7, e33872 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Liu, J. et al. Impaired adult myelination in the prefrontal cortex of socially isolated mice. Nat. Neurosci. 15, 1621–1623 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Morel, E. et al. The cellular prion protein PrP(c) is involved in the proliferation of epithelial cells and in the distribution of junction-associated proteins. PLoS ONE 3, e3000 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Labonte, B. et al. Sex-specific transcriptional signatures in human depression. Nat. Med. 23, 1102–1111 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dumais, A. et al. Risk factors for suicide completion in major depression: a case–control study of impulsive and aggressive behaviors in men. Am. J. Psychiatry 162, 2116–2124 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2017).

  53. Benaglia, T., Chauveau, D., Hunter, D. R. & Young, D. S. mixtools: an R Package for analyzing mixture models. J. Stat. Soft. 32, 29 (2009).

    Article  Google Scholar 

  54. Paradis, E., Claude, J. & Strimmer, K. APE: analyses of phylogenetics and evolution in R language. Bioinformatics 20, 289–290 (2004).

    Article  CAS  PubMed  Google Scholar 

  55. Hawrylycz, M. J. et al. An anatomically comprehensive atlas of the adult human brain transcriptome. Nature 489, 391–399 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lake, B. B. et al. Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain. Science 352, 1586–1590 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bates, D., Mächler, M., Bolker, B. & Walker, S. Fitting linear mixed-effects models using lme4. J. Stat. Soft. 67, 48 (2015).

    Article  Google Scholar 

  58. Kuznetsova, A., Brockhoff, P. B. & Christensen, R. H. B. lmerTest Package: tests in linear mixed effects models. J. Stat. Soft. 82, 26 (2017).

    Article  Google Scholar 

  59. Lutz, P. E. et al. Association of a history of child abuse with impaired myelination in the anterior cingulate cortex: convergent epigenetic, transcriptional, and morphological evidence. Am. J. Psychiatry 174, 1185–1194 (2017).

    Article  PubMed  Google Scholar 

  60. Bayega, A. et al. in Gene Expression Analysis: Methods and Protocols (eds Raghavachari, N. & Garcia-Reyero, N.) 121–147 (Springer New York, 2018).

  61. Spurgeon, S. L., Jones, R. C. & Ramakrishnan, R. High throughput gene expression measurement with real time PCR in a microfluidic dynamic array. PLoS ONE 3, e1662 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Ximerakis, M. et al. Single-cell transcriptomic profiling of the aging mouse brain. Nat. Neurosci. 22, 1696–1708 (2019).

    Article  CAS  PubMed  Google Scholar 

  63. Gong, T. & Szustakowski, J. D. DeconRNASeq: a statistical framework for deconvolution of heterogeneous tissue samples based on mRNA-Seq data. Bioinformatics 29, 1083–1085 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

G.T. holds a Canada Research Chair (Tier 1) and a NARSAD Distinguished Investigator Award. He is supported by grants from the Canadian Institute of Health Research (CIHR) (FDN148374 and EGM141899). We acknowledge the expert help of the Douglas–Bell Canada Brain Bank staff (J. Prud’homme, M. Bouchouka and A. Baccichet), and H. Djambazian at the MUGQIC. The work was also supported by CFI projects 32557 and 33408 (to J.R.). The Douglas–Bell Canada Brain Bank is supported in part by funding from the Canada First Research Excellence Fund, awarded to McGill University for the Healthy Brains for Healthy Lives project, and from the Fonds de recherche du Québec–Santé (FRQS) through the Quebec Network on Suicide, Mood Disorders and Related Disorders. The present study used the services of the Molecular and Cellular Microscopy Platform (MCMP) at the Douglas Institute.

Author information

Authors and Affiliations

Authors

Contributions

C.N. conceptualized, performed experiments and wrote the manuscript. M.M. performed experiments, bioinformatics and wrote the manuscript. A.T., V.Y., M.A.D. and Y.C.W. performed experiments and wrote the manuscript. M.S., K.P., J.-F.T., S.J.T. and P.P. contributed to data analyses and reviewed the manuscript. N.M. contributed to tissue processing, data interpretation and manuscript preparation. J.R. provided technical single-cell expertise and experimental support, and aided in manuscript preparation. G.T. provided general oversight of the study, including in experimental design, data interpretation and manuscript preparation.

Corresponding author

Correspondence to Gustavo Turecki.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Ronald Duman (deceased), Matthew Girgenti, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Methods and Supplementary Figs. 1–14.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–52.

Supplementary Software

Supplementart_R_Script_1.R for analyzing high-throughput qPCR data and Supplementary_R_Script_2.R for generating P values for CCInx interactions.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nagy, C., Maitra, M., Tanti, A. et al. Single-nucleus transcriptomics of the prefrontal cortex in major depressive disorder implicates oligodendrocyte precursor cells and excitatory neurons. Nat Neurosci 23, 771–781 (2020). https://doi.org/10.1038/s41593-020-0621-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-020-0621-y

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