Targeted single-cell RNA sequencing of transcription factors enhances the identification of cell types and trajectories

  1. M. Zameel Cader1
  1. 1Translational Molecular Neuroscience Group, Weatherall Institute of Molecular Medicine, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DS, United Kingdom;
  2. 2Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, United Kingdom;
  3. 3UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, United Kingdom;
  4. 4Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, United Kingdom;
  5. 5Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, United Kingdom;
  6. 6Centre for Stem Cell Systems, Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria 3010, Australia;
  7. 7The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia;
  8. 8University of Melbourne, Department of Medical Biology, Parkville, Victoria 3052, Australia
  • Corresponding authors: zameel.cader{at}ndcn.ox.ac.uk, bowden.r{at}wehi.edu.au, michael.clark{at}unimelb.edu.au
  • Abstract

    Single-cell RNA sequencing (scRNA-seq) is a widely used method for identifying cell types and trajectories in biologically heterogeneous samples, but it is limited in its detection and quantification of lowly expressed genes. This results in missing important biological signals, such as the expression of key transcription factors (TFs) driving cellular differentiation. We show that targeted sequencing of ∼1000 TFs (scCapture-seq) in iPSC-derived neuronal cultures greatly improves the biological information garnered from scRNA-seq. Increased TF resolution enhanced cell type identification, developmental trajectories, and gene regulatory networks. This allowed us to resolve differences among neuronal populations, which were generated in two different laboratories using the same differentiation protocol. ScCapture-seq improved TF-gene regulatory network inference and thus identified divergent patterns of neurogenesis into either excitatory cortical neurons or inhibitory interneurons. Furthermore, scCapture-seq revealed a role for of retinoic acid signaling in the developmental divergence between these different neuronal populations. Our results show that TF targeting improves the characterization of human cellular models and allows identification of the essential differences between cellular populations, which would otherwise be missed in traditional scRNA-seq. scCapture-seq TF targeting represents a cost-effective enhancement of scRNA-seq, which could be broadly applied to improve scRNA-seq resolution.

    Footnotes

    • 9 Co-first authors.

    • [Supplemental material is available for this article.]

    • Article published online before print. Article, supplemental material, and publication date are at https://www.genome.org/cgi/doi/10.1101/gr.273961.120.

    • Freely available online through the Genome Research Open Access option.

    • Received November 5, 2020.
    • Accepted March 23, 2021.

    This article, published in Genome Research, is available under a Creative Commons License (Attribution 4.0 International), as described at http://creativecommons.org/licenses/by/4.0/.

    | Table of Contents
    OPEN ACCESS ARTICLE

    Preprint Server