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.

Electrophysiological, transcriptomic and morphologic profiling of single neurons using Patch-seq

Abstract

Despite the importance of the mammalian neocortex for complex cognitive processes, we still lack a comprehensive description of its cellular components. To improve the classification of neuronal cell types and the functional characterization of single neurons, we present Patch-seq, a method that combines whole-cell electrophysiological patch-clamp recordings, single-cell RNA-sequencing and morphological characterization. Following electrophysiological characterization, cell contents are aspirated through the patch-clamp pipette and prepared for RNA-sequencing. Using this approach, we generate electrophysiological and molecular profiles of 58 neocortical cells and show that gene expression patterns can be used to infer the morphological and physiological properties such as axonal arborization and action potential amplitude of individual neurons. Our results shed light on the molecular underpinnings of neuronal diversity and suggest that Patch-seq can facilitate the classification of cell types in the nervous system.

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

Figure 1: Two morphologically and electrophysiologically distinct neuronal classes in neocortical layer 1.
Figure 2: Single-neuron transcriptome profiles predict cell type and electrophysiological properties.
Figure 3: Differential gene expression analysis reveals novel markers for L1 interneuron classes.

Similar content being viewed by others

Accession codes

Primary accessions

ArrayExpress

References

  1. Cajal, S.R., Pasik, P. & Pasik, T. Texture of the Nervous System of Man and the Vertebrates (Springer, 2002).

  2. Ascoli, G.A. et al. Petilla Interneuron Nomenclature Group. Petilla terminology: nomenclature of features of GABAergic interneurons of the cerebral cortex. Nat. Rev. Neurosci. 9, 557–568 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Burkhalter, A. Many specialists for suppressing cortical excitation. Front. Neurosci. 2, 155–167 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jiang, X. et al. Principles of connectivity among morphologically defined cell types in adult neocortex. Science 350, aac9462 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Connors, B.W. & Gutnick, M.J. Intrinsic firing patterns of diverse neocortical neurons. Trends Neurosci. 13, 99–104 (1990).

    Article  CAS  PubMed  Google Scholar 

  6. Neher, E. & Sakmann, B. Single-channel currents recorded from membrane of denervated frog muscle fibres. Nature 260, 799–802 (1976).

    Article  CAS  PubMed  Google Scholar 

  7. Tang, F. et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009).

    CAS  PubMed  Google Scholar 

  8. Sandberg, R. Entering the era of single-cell transcriptomics in biology and medicine. Nat. Methods 11, 22–24 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Fishell, G. & Heintz, N. The neuron identity problem: form meets function. Neuron 80, 602–612 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Jaitin, D.A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Darmanis, S. et al. A survey of human brain transcriptome diversity at the single cell level. Proc. Natl. Acad. Sci. USA 112, 7285–7290 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zeisel, A. et al. Brain structure. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science 347, 1138–1142 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Sucher, N.J., Deitcher, D.L., Baro, D.J., Warrick, R.M. & Guenther, E. Genes and channels: patch/voltage-clamp analysis and single-cell RT-PCR. Cell Tissue Res. 302, 295–307 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Toledo-Rodriguez, M. & Markram, H. Single-cell RT-PCR, a technique to decipher the electrical, anatomical, and genetic determinants of neuronal diversity. Methods Mol. Biol. 1183, 143–158 (2014).

    Article  PubMed  Google Scholar 

  15. Subkhankulova, T., Yano, K., Robinson, H.P. & Livesey, F.J. Grouping and classifying electrophysiologically-defined classes of neocortical neurons by single cell, whole-genome expression profiling. Front. Mol. Neurosci. 3, 10 (2010).

    PubMed  PubMed Central  Google Scholar 

  16. Qiu, S. et al. Single-neuron RNA-Seq: technical feasibility and reproducibility. Front. Genet. 3, 124 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. McGinley, M.J. et al. Waking state: rapid variations modulate neural and behavioral responses. Neuron 87, 1143–1161 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Chan, C.H. et al. Emx1 is a marker for pyramidal neurons of the cerebral cortex. Cereb. Cortex 11, 1191–1198 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Fremeau, R.T. Jr. et al. The expression of vesicular glutamate transporters defines two classes of excitatory synapse. Neuron 31, 247–260 (2001).

    Article  CAS  PubMed  Google Scholar 

  21. Marshak, D.R. S100 beta as a neurotrophic factor. Prog. Brain Res. 86, 169–181 (1990).

    Article  CAS  PubMed  Google Scholar 

  22. Bignami, A., Eng, L.F., Dahl, D. & Uyeda, C.T. Localization of the glial fibrillary acidic protein in astrocytes by immunofluorescence. Brain Res. 43, 429–435 (1972).

    Article  CAS  PubMed  Google Scholar 

  23. Stühmer, T., Anderson, S.A., Ekker, M. & Rubenstein, J.L. Ectopic expression of the Dlx genes induces glutamic acid decarboxylase and Dlx expression. Development 129, 245–252 (2002).

    PubMed  Google Scholar 

  24. Alcántara, S. et al. Regional and cellular patterns of reelin mRNA expression in the forebrain of the developing and adult mouse. J. Neurosci. 18, 7779–7799 (1998).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Miyoshi, G. et al. Genetic fate mapping reveals that the caudal ganglionic eminence produces a large and diverse population of superficial cortical interneurons. J. Neurosci. 30, 1582–1594 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ma, J., Yao, X.H., Fu, Y. & Yu, Y.C. Development of layer 1 neurons in the mouse neocortex. Cereb. Cortex 24, 2604–2618 (2014).

    Article  PubMed  Google Scholar 

  27. Kharchenko, P.V., Silberstein, L. & Scadden, D.T. Bayesian approach to single-cell differential expression analysis. Nat. Methods 11, 740–742 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jiang, X., Wang, G., Lee, A.J., Stornetta, R.L. & Zhu, J.J. The organization of two new cortical interneuronal circuits. Nat. Neurosci. 16, 210–218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Oláh, S. et al. Regulation of cortical microcircuits by unitary GABA-mediated volume transmission. Nature 461, 1278–1281 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Kalashnikova, E. et al. SynDIG1: an activity-regulated, AMPA- receptor-interacting transmembrane protein that regulates excitatory synapse development. Neuron 65, 80–93 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Macintyre, G. et al. Association of NPAS3 exonic variation with schizophrenia. Schizophr. Res. 120, 143–149 (2010).

    Article  PubMed  Google Scholar 

  32. Stanco, A. et al. NPAS1 represses the generation of specific subtypes of cortical interneurons. Neuron 84, 940–953 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lin, L. et al. DPP6 regulation of dendritic morphogenesis impacts hippocampal synaptic development. Nat. Commun. 4, 2270 (2013).

    Article  PubMed  Google Scholar 

  34. Brose, N. For better or for worse: complexins regulate SNARE function and vesicle fusion. Traffic 9, 1403–1413 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Chao, H.T. et al. Dysfunction in GABA signalling mediates autism-like stereotypies and Rett syndrome phenotypes. Nature 468, 263–269 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ting, J.T., Daigle, T.L., Chen, Q. & Feng, G. Acute brain slice methods for adult and aging animals: application of targeted patch clamp analysis and optogenetics. Methods Mol. Biol. 1183, 221–242 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Sucher, N.J. & Deitcher, D.L. PCR and patch-clamp analysis of single neurons. Neuron 14, 1095–1100 (1995).

    Article  CAS  PubMed  Google Scholar 

  38. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Picelli, S. et al. Tn5 transposase and tagmentation procedures for massively scaled sequencing projects. Genome Res. 24, 2033–2040 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Ramsköld, D., Wang, E.T., Burge, C.B. & Sandberg, R. An abundance of ubiquitously expressed genes revealed by tissue transcriptome sequence data. PLoS Comput. Biol. 5, e1000598 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Brennecke, P. et al. Accounting for technical noise in single-cell RNA-seq experiments. Nat. Methods 10, 1093–1095 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Friedman, J., Hastie, T. & Tibshirani, R. Regularization Paths for Generalized Linear Models via Coordinate Descent. J. Stat. Softw. 33, 1–22 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Clopper, C. & Pearson, E.S. The use of confidence or fiducial limits illustrated in the case of the binomial. Biometrika 26, 404–413 (1934).

    Article  Google Scholar 

Download references

Acknowledgements

We thank A. Morgan for technical assistance. This study was supported by grants DP1EY023176, P30EY002520, T32EY07001,and DP1OD008301 to A.S.T.; grants from the Swedish Research Council and the Swedish Foundation for Strategic Research (FFL4) to R.S.; grant R01MH103108 to A.S.T. and K.F.T.; grant R01NS062829 to K.F.T.; the McKnight Scholar Award to A.S.T.; and the Arnold and Mabel Beckman Foundation Young Investigator Award to A.S.T. C.R.C. was supported by grants F30MH095440, T32GM007330 and T32EB006350. M.B. and P.B. were supported by the Deutsche Forschungsgemeinschaft (DFG, EXC 307) and the German Federal Ministry of Education and Research (BMBF; BCCN Tübingen, FKZ 01GQ1002).

Author information

Authors and Affiliations

Authors

Contributions

C.R.C. collected RNA samples, generated cDNA libraries, assisted with analysis and drafted the manuscript. A.P. performed the computational analyses of RNA-seq data. X.J. performed the ex vivo patch-clamp experiments, reconstructed neuronal morphologies and analyzed electrophysiological properties of the neurons. P.B. built the automated cell type classifier and generalized linear models. Q.D. and M.Y. generated cDNA and sequencing libraries. J.R. and S.S. performed the in vivo patch-clamp experiments. M.B. supervised the machine learning analysis. A.S.T., R.S. and K.F.T. supervised all experiments and analyses. All authors contributed to writing the paper.

Corresponding authors

Correspondence to Rickard Sandberg or Andreas S Tolias.

Ethics declarations

Competing interests

R.S. has developed and patented Smart-seq2 and licensed that technology to Clontech, a Takara Bio Company.

Integrated supplementary information

Supplementary Figure 1 Optimization of Patch-seq protocol.

Pilot experiments were carried out to test the effect of various protocol modifications on cDNA yield, including addition of RNase inhibitor to the intracellular solution (a), silanization of glass capillaries (b), concentration of dNTPs in lysis buffer (c), extraction of cytoplasm only v. cytoplasm and nucleus (d), pipette tip size (e) (tip size inversely proportional to resistance, line represents linear regression), and volume of intracellular solution in patch pipette (f) (20 μl total reaction volume, RT-PCR performed on whole brain cDNA template).

Supplementary Figure 2 Modified RNase-free intracellular solution does not affect health of patch-clamp-recorded neurons.

Membrane potential of neurons patched with RNase-free modified intracellular solution for Patch-seq recordings. Data represent mean ± SE (n=3 neurons).

Supplementary Figure 3 Quality control of cDNA libraries.

(a) Average size and concentration (from 300-9,000 bp) of all collected samples. Only samples with more than 200 pg/μl and an average size greater than 1,500 bp were sequenced (criteria denoted by dashed lines). Libraries generated from patched cells generally contained more (b) and higher quality, full-length (c) cDNA compared to negative controls in which no cell was patched. Distributions of concentrations (d) and average sizes (e) of cDNA libraries generated from ex vivo patched cells. Dashed lines in (b-e) indicate threshold criteria used for sequencing. (f) Pearson correlation between concentration of cDNA and number of genes detected for all sequenced ex vivo cells.

Supplementary Figure 4 Mapping statistics of Patch-seq and standard Smart-seq2 libraries.

(a) Percentage of sequenced reads that aligned uniquely to the respective assembly for patched single neurons and dissociated HEK293T cells picked and sequenced according to standard Smart-seq2 protocol (Picelli et al. 2013). (b) Percentage of uniquely aligned reads that overlap annotated RefSeq exons, introns or between gene annotations. This comparison indicates that the libraries generated from patched single neurons have similar qualities as standard Smart-seq2 libraries generated from lysed whole cells.

Supplementary Figure 5 Dimension reductions and marker expression in interneurons, pyramidal neurons and astrocyte.

(a) Principal component analyses (PCA) of the expression profiles of 16,000 genes detected in any of the patched cells. Each cell was projected onto a two-dimensional space drawn by the first two principal components. (b) Two-dimensional representation of cells using t-SNE on the PCA space, as in (a) but considering the first 28 principal components. Note that single outlier cells cannot be well captured in t-SNE representations as a consequence of the neighborhood analyses of the closest 20 cells (perplexity parameter). Therefore, the distinct gene expression observed in the astrocyte in the PCA (a) is not well captured in the t-SNE map. (c) Boxplots that show marker gene expression for interneurons, pyramidal neurons and the astrocyte. The mean and median are represented by the circle and black line, respectively. The edges of the box represent the 25th and 75th percentiles. Outlier cells are marked with grey dots, and the whiskers extend to extreme data points not considered outliers.

Supplementary Figure 6 Marker gene expression overlaid onto t-SNE map.

Marker gene expression overlaid onto the two-dimensional t-SNE representation (from Fig. 2d,e). Colors according to log2- transformed gene expression values.

Supplementary Figure 7 Clustering of random samples of L1 interneurons.

Investigating the smallest number of patched cells needed to identify the two L1 interneuron cell types with unbiased clustering of gene expression profiles. (a) Affinity propagation was used to cluster random subsets of L1 interneurons (ex vivo cells, n=46) at decreasing numbers of cells. After each subsampling and clustering, the cell assignments were compared to the assignment based on all cells (Fig. 2d) and the accuracy score calculated (i.e. the number of correctly assigned cells over all subsampled cells). Accuracy scores from the 250 iterations per step are shown as standard boxplots, with median and mean as a line and circle, respectively. Importantly, even with random samples of 31 cells we robustly captured the two cell types (median accuracy=90%). Sampling fewer cells, in particular below 26 cells, often failed to distinguish the two cell types in an unbiased manner. (b) Showing the percentage of iterations where the affinity propagation clustering resulted in one, or two or three clusters, with the subsampling of decreasing numbers of L1 interneurons. The decline in performance at lower cell numbers was mainly caused by an inability of the affinity propagation algorithm to find two clusters, and instead reporting only one cluster.

Supplementary Figure 8 Expression differences in interneurons patch-clamp-recorded ex vivo and in vivo.

(a) Coloring the t-SNE map (from Fig. 2d,e) according to ex vivo and in vivo patched neurons. (b) Heat map showing the top 17 genes that separate ex vivo and in vivo cells The genes were identified using SCDE and all have absolute values of Z-score above 1.96 (genes with asterisk have corrected absolute Z-score above 1.96). The heat map shows an increased expression of genes ex vivo relating to stress induction (including Fos, Fosb, Junb) and other genes with differential expression.

Supplementary Figure 9 Overlaying electrophysiological properties of cells onto the two-dimensional t-SNE map.

Coloring of cells in the t-SNE space according to their electrophysiological properties. (a) Identical to Fig. 2e, showing cell type assignments onto the t-SNE map, included to ease the comparison to panels b-l. (b-j) Cells colored according to continuous electrophysiological features, scaled by the highest and lowest measurement onto the indicated colorbar. (k-l) Cells colored based on their displayed spiking patterns during recordings. Grey squares show in vivo patched cells.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 (PDF 2826 kb)

Supplementary Table 1

Mapping statistics of genes aligned uniquely, multimapping or unmapped. (XLSX 60 kb)

Supplementary Table 2

List with genes ranked according to biological variation. (XLSX 353 kb)

Supplementary Table 3

Genes used in regularized GLMs for predicting cell class and physiological properties (XLSX 39 kb)

Supplementary Table 4

Analyses of differential gene expression between interneurons of different cell type or electrophysiological properties. (XLSX 4558 kb)

Supplementary Table 5

Results obtained using as background the genes expressed across L1 interneurons (~ 6,300) and comparing against the top 200 differentially expressed genes (up-regulated) within cluster B (SBCs) (XLSX 80 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cadwell, C., Palasantza, A., Jiang, X. et al. Electrophysiological, transcriptomic and morphologic profiling of single neurons using Patch-seq. Nat Biotechnol 34, 199–203 (2016). https://doi.org/10.1038/nbt.3445

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3445

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