Skip to main content

Single-cell genomics: coming of age

Single-cell genomics is the study of the individuality of cells using omics approaches. Although young, the field has now entered its teenage years and is beginning to show clear signs of maturity. Its origins can be traced back to pioneering experiments that allowed the detection of gene expression in single cells by microarrays (reviewed in [1]). However, it was with the emergence of “next-generation” DNA sequencing that single-cell genomics really took off [24]. Although initial experiments were modest in size and resulted in noisy and incomplete data, they immediately revealed the great potential for biological discoveries. It soon became clear that the substantial technical and biological variability required data from many single cells in order to allow meaningful data mining and interpretation of the data [5]. Thus, the following years were spent pursuing a few lines of development: improvements in the accuracy and scope of single-cell methods and increasing throughput and reducing cost. Today, we are in a position to routinely measure gene expression in tens of thousands of single cells with high accuracy in terms of quantification of gene expression (albeit sensitivity in terms of detection of mRNAs varies significantly depending on protocol and sequencing depth). The costs are at least manageable and continue to decrease.

While single-cell RNA-seq is now mature and almost routine, technological development has shifted to other modalities: DNA, protein, chromatin modifications, and more. Single-cell whole-genome DNA sequencing is challenging because loss of material causes dropouts in the sequence and because sequencing errors are difficult to distinguish from real mutations. Despite these challenges, single human cortical neurons have been used to reconstruct lineages based on somatic mutations that had accumulated during development [6]. Similarly, clonal evolution within solid tumors can be revealed by detecting somatic copy number variations in single cells (reviewed in [7]).

Another trend is the extension of single-cell analysis to measure epigenetic states such as DNA accessibility [810], methylation [11], and chromosome conformation [12]. Generally these methods pose similar challenges to DNA sequencing but offer access to pure cellular epigenetic states that are simply inaccessible by bulk methods.

Single-cell protein analysis occupies a different niche, where smaller numbers of proteins can be analyzed but in very large numbers of cells, classically using fluorescence-activated cell sorting (FACS) for up to eight targets but more recently with mass cytometry targeting up to hundreds of proteins [13]. A limiting factor for protein analysis remains the requirement for high-quality affinity reagents such as antibodies.

Finally, a recent development (but see [14]) is the combination of methods to simultaneously measure two or more modalities in single cells. For example, genome and transcriptome [15, 16], transcriptome and methylome [17, 18], and RNA and protein [19]. In the near future, such experiments will be able to link the phenotypes of single cells evolving in tumors to their genotypes.

Due to the speed with which single-cell genomics technologies are evolving, computational analysis methods are racing to keep up. Statistical and computational methods are at the heart of single-cell genomics and are critical to extracting meaningful information and biology from the data. Much work has focused on transcriptomic data analysis (e.g., reviewed in [20]) and in this special issue of Genome Biology there are examples of areas that benefit from bespoke computational approaches at the levels of both cells and genes. In terms of individual genes, a method to define significant differences in the cell-to-cell variation in gene expression (as opposed to mean expression levels) is reported [21] and one paper addresses expression states of long noncoding RNAs [22]. In terms of cell-to-cell variation at the DNA level, there is clearly tremendous scope for computational method innovation in the area of tumor heterogeneity, addressed by Beerenwinkel and colleagues [23], and Markowetz and Ross [24] in this issue.

Recent applications

Single-cell RNA sequencing has had a profound impact on our understanding of neuronal and hematopoietic cell types, as well as the immune system. Examples of novel insights in immunity include a window on to an unexpected plethora of dendritic cells in mouse immunity [25] and new regulators and subpopulations of CD4+ T cells [2628]. In hematopoiesis, much single-cell transcriptomics work has focused on hematopoetic stem cells and the single-cell perspective has provided resolution of proliferation phenotypes [2931]. A broader view of early specification of hematopoietic cell types was recently provided by Paul et al. [32]. Mead and colleagues [33] provide new insights into the erythroid–myeloid decision in this special issue.

While these publications all focus on mouse as a model, the unbiased nature of single-cell RNA sequencing provides great discovery potential in less-well-studied animals. An example of this is the profiling of platelets (thrombocytes) from hematopoietic stem cells in zebrafish by Macaulay et al. [34]. In this issue, Pearson and Molinaro profile single cells in planarian regeneration [35]. Looking to the future, this type of approach can be expanded to comparative studies of many organisms across the animal kingdom in order to gain insight into the evolution of cell types.

The applicability of single-cell transcriptomics to nonadherent cells, such as those of hematopoiesis and immunity, is perhaps not surprising: these cells naturally exist as individual cells and remain stable after single-cell capture by FACS or in microfluidic devices. In the area of neurobiology and neuronal cell populations, the success of single-cell RNA sequencing is more surprising as these cells are bound up within networks of adherent junctions. Recently, comprehensive maps of cell types and subtypes have been produced for a number of key brain regions, including developing and adult cerebral cortex, and the day will come when we will have a full catalog of molecularly defined cell types in the whole nervous system. A particularly appealing application of such a reference atlas is in the use of human cerebral organoids to model human brain (which is otherwise inaccessible) in development and disease [36]. The fact that novel cell states, cell populations, and factors have been validated in this domain bodes well for a broader remit of single-cell transcriptomics to solid organs and tissues.

The DNA dimension, i.e., tracking mutations, copy number variations, and chromosomal aberrations at the single-cell level, has been important in both somatic cell populations such as neurons, as well as in cancer. In this issue, Park and colleagues show how single-cell dissection of tumor heterogeneity can translate directly into new combinatorial therapies in a xenograft model [37].

Future prospects

Gazing into our crystal ball, it is easy to predict an ever-increasing role for single-cell genomics in discovery science, translational applications, and even ecology. The major driver of the single-cell genomics revolution is the step change in resolution of DNA and epigenetic and RNA sequencing down to the level of an individual cell. Since the cell is the basic building block of an organism, sequencing each cell in isolation provides information that is fundamentally different from genomic data that relates to ensembles of cells.

In terms of single-cell transcriptomics, the RNA content of a cell is deeply informative about its phenotype and function. This technique is so powerful and informative that it is likely that the community will ultimately map all mammalian organs, tissues, and cell types at single-cell resolution. A comprehensive resource such as this, effectively a “human cell atlas”, would be a tremendously useful and unique reference data set for biology and medicine.

Like many previous waves of biotechnology, single-cell genomics started in academia and basic research but is now set to move into pharma and the clinic. Once an atlas of human cell types is available, any diseased tissue can be compared with it. Cancer, in particular, the prototypical single-cell disease, will be particularly apt for a single-cell analysis overhaul. Diagnostic assays, which are currently based on crude bulk methods, will be tremendously more powerful once they are brought down to the level of the individual transformed cell, in the context of its surrounding tissue, with cell-type specificity and a full understanding of somatic mutations.

We are excited to be part of a community that has already achieved a lot, as showcased in this special issue, yet clearly still has a long and interesting journey ahead of it.

Abbreviations

FACS:

fluorescence-activated cell sorting

References

  1. Tang F, Lao K, Surani MA. Development and applications of single-cell transcriptome analysis. Nat Methods. 2011;8:S6–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. 2009;6:377–82.

    Article  CAS  PubMed  Google Scholar 

  3. Islam S, Kjallquist U, Moliner A, Zajac P, Fan JB, Lonnerberg P, et al. Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq. Genome Res. 2011;21:1160–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ramskold D, Luo S, Wang YC, Li R, Deng Q, Faridani OR, et al. Full-length mRNA-seq from single-cell levels of RNA and individual circulating tumor cells. Nat Biotechnol. 2012;30:777–82.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Grun D, van Oudenaarden A. Design and analysis of single-cell sequencing experiments. Cell. 2015;163:799–810.

    Article  CAS  PubMed  Google Scholar 

  6. Lodato MA, Woodworth MB, Lee S, Evrony GD, Mehta BK, Karger A, et al. Somatic mutation in single human neurons tracks developmental and transcriptional history. Science. 2015;350:94–8.

    Article  CAS  PubMed  Google Scholar 

  7. Navin NE. The first five years of single-cell cancer genomics and beyond. Genome Res. 2015;25:1499–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, Snyder MP, et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature. 2015;523:486–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cusanovich DA, Daza R, Adey A, Pliner HA, Christiansen L, Gunderson KL, et al. Epigenetics. Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing. Science. 2015;348:910–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Jin W, Tang Q, Wan M, Cui K, Zhang Y, Ren G, et al. Genome-wide detection of DNase I hypersensitive sites in single cells and FFPE tissue samples. Nature. 2015;528:142–6.

    CAS  PubMed  Google Scholar 

  11. Smallwood SA, Lee HJ, Angermueller C, Krueger F, Saadeh H, Peat J, et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat Methods. 2014;11:817–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nagano T, Lubling Y, Stevens TJ, Schoenfelder S, Yaffe E, Dean W, et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature. 2013;502:59–64.

    Article  CAS  PubMed  Google Scholar 

  13. Bendall SC, Simonds EF, Qiu P, Amir e-AD, Krutzik PO, Finck R, et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science. 2011;332:687–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Klein CA, Seidl S, Petat-Dutter K, Offner S, Geigl JB, Schmidt-Kittler O, et al. Combined transcriptome and genome analysis of single micrometastatic cells. Nat Biotechnol. 2002;20:387–92.

    Article  CAS  PubMed  Google Scholar 

  15. Macaulay IC, Haerty W, Kumar P, Li YI, Hu TX, Teng MJ, et al. G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nat Methods. 2015;12:519–22.

    Article  CAS  PubMed  Google Scholar 

  16. Dey SS, Kester L, Spanjaard B, Bienko M, van Oudenaarden A. Integrated genome and transcriptome sequencing of the same cell. Nat Biotechnol. 2015;33:285–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Moroz LL, Kohn AB. Single-neuron transcriptome and methylome sequencing for epigenomic analysis of aging. Methods Mol Biol. 2013;1048:323–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Angermueller C, Clark SJ, Lee HJ, Macaulay IC, Teng MJ, Hu TX, et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat Methods. 2016;13:229–32.

    Article  CAS  PubMed  Google Scholar 

  19. Darmanis S, Gallant CJ, Marinescu VD, Niklasson M, Segerman A, Flamourakis G, et al. Simultaneous multiplexed measurement of RNA and proteins in single cells. Cell Rep. 2016;14:380–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Stegle O, Teichmann SA, Marioni JC. Computational and analytical challenges in single-cell transcriptomics. Nat Rev Genet. 2015;16:133–45.

    Article  CAS  PubMed  Google Scholar 

  21. Vallejos CA, Richardson S, Marioni JC. Beyond comparisons of means: understanding changes in gene expression at the single-cell level. Genome Biol. 2016;17:70.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Liu SJ, Nowakowski TJ, Pollen AA, Lui JH, Horlbeck MA, Attenello FJ, et al. Single-cell analysis of long non-coding RNAs in the developing human neocortex. Genome Biol. 2016;17:67.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Jahn K, Kuipers J, Beerenwinkel N. Tree inference for single-cell data. Genome Biol. 2016. doi:10.1186/s13059-016-0936-x.

    PubMed  Google Scholar 

  24. Ross EM, Markowetz F. OncoNEM: inferring tumor evolution from single-cell sequencing data. Genome Biol. 2016;17:69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jaitin DA, Kenigsberg E, Keren-Shaul H, Elefant N, Paul F, Zaretsky I, et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science. 2014;343:776–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mahata B, Zhang X, Kolodziejczyk AA, Proserpio V, Haim-Vilmovsky L, Taylor AE, et al. Single-cell RNA sequencing reveals T helper cells synthesizing steroids de novo to contribute to immune homeostasis. Cell Rep. 2014;7:1130–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Proserpio V, Piccolo A, Haim-Vilmovsky L, Kar G, Lonnberg T, Svensson V, et al. Single cell analysis of CD4+ T cell differentiation reveals three major cell states and progressive acceleration of proliferation. Genome Biol. 2016. doi:10.1186/s13059-016-0957-5.

    Google Scholar 

  28. Gaublomme JT, Yosef N, Lee Y, Gertner RS, Yang LV, Wu C, et al. Single-cell genomics unveils critical regulators of Th17 cell pathogenicity. Cell. 2015;163:1400–12.

    Article  CAS  PubMed  Google Scholar 

  29. Kowalczyk MS, Tirosh I, Heckl D, Rao TN, Dixit A, Haas BJ, et al. Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome Res. 2015;25:1860–72.

    Article  PubMed  Google Scholar 

  30. Tsang JC, Yu Y, Burke S, Buettner F, Wang C, Kolodziejczyk AA, et al. Single-cell transcriptomic reconstruction reveals cell cycle and multi-lineage differentiation defects in Bcl11a-deficient hematopoietic stem cells. Genome Biol. 2015;16:178.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Wilson NK, Kent DG, Buettner F, Shehata M, Macaulay IC, Calero-Nieto FJ, et al. Combined single-cell functional and gene expression analysis resolves heterogeneity within stem cell populations. Cell Stem Cell. 2015;16:712–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Paul F, Arkin Y, Giladi A, Jaitin DA, Kenigsberg E, Keren-Shaul H, et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell. 2015;163:1663–77.

    Article  CAS  PubMed  Google Scholar 

  33. Psaila B, Barkas N, Iskander D, Roy A, Anderson S, Ashley N, et al. Single-cell profiling of human megakaryocyte-erythroid progenitors identifies distinct megakaryocyte and erythroid differentiation pathways. Genome Biol. 2016. doi:10.1186/s13059-016-0939-7.

    PubMed  PubMed Central  Google Scholar 

  34. Macaulay IC, Svensson V, Labalette C, Ferreira L, Hamey F, Voet T, et al. Single-cell RNA-sequencing reveals a continuous spectrum of differentiation in hematopoietic cells. Cell Rep. 2016;14:966–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Molinaro AM, Pearson BJ. In silico lineage tracing through single cell transcriptomics identifies a neural stem cell population in planarians. Genome Biol. 2016. doi:10.1186/s13059-016-0937-9.

    PubMed  Google Scholar 

  36. Camp JG, Badsha F, Florio M, Kanton S, Gerber T, Wilsch-Brauninger M, et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci U S A. 2015;112:15672–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kim KT, Lee HW, Lee HO, Song HJ, Jeong DE, Shin S, et al. Application of single-cell RNA-sequencing in optimizing a combinatorial therapeutic strategy in metastatic renal cell carcinoma. Genome Biol. 2016. doi:10.1186/s13059-016-0945-9.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Sten Linnarsson or Sarah A. Teichmann.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Both authors read and approved the final manuscript.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Linnarsson, S., Teichmann, S.A. Single-cell genomics: coming of age. Genome Biol 17, 97 (2016). https://doi.org/10.1186/s13059-016-0960-x

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/s13059-016-0960-x