5,215
Views
20
CrossRef citations to date
0
Altmetric
Posterior Segment

Understanding the complexity of retina and pluripotent stem cell derived retinal organoids with single cell RNA sequencing: current progress, remaining challenges and future prospective

, , , &
Pages 385-396 | Received 13 Sep 2019, Accepted 22 Oct 2019, Published online: 03 Feb 2020

ABSTRACT

Single-cell sequencing technologies have emerged as a revolutionary tool with transformative new methods to profile genetic, epigenetic, spatial, and lineage information in individual cells. Single-cell RNA sequencing (scRNA-Seq) allows researchers to collect large datasets detailing the transcriptomes of individual cells in space and time and is increasingly being applied to reveal cellular heterogeneity in retinal development, normal physiology, and disease, and provide new insights into cell-type specific markers and signaling pathways. In recent years, scRNA-Seq datasets have been generated from retinal tissue and pluripotent stem cell-derived retinal organoids. Their cross-comparison enables staging of retinal organoids, identification of specific cells in developing and adult human neural retina and provides deeper insights into cell-type sub-specification and geographical differences. In this article, we review the recent rapid progress in scRNA-Seq analyses of retina and retinal organoids, the questions that remain unanswered and the technical challenges that need to be overcome to achieve consistent results that reflect the complexity, functionality, and interactions of all retinal cell types.

Introduction

Single-cell RNA sequencing (scRNA-Seq) is a powerful tool for exploring the transcriptional heterogeneity of large cell populations by measuring gene transcription in each individual cell. Elucidating the cellular heterogeneity within a biological system is an essential requisite for understanding development and evolution, how a biological system is homeostatically regulated, and responds to external stimuli. Today, with the rapid development of the single-cell-based technologies it is possible to determine the mRNA levels of several thousand transcripts in thousands of cells as well as map their spatial distribution through spatial transcriptomics techniques.Citation1 The first paper which described the feasibility of single cell-RNA sequencing was published 10 years ago where the authors profiled only eight cells.Citation2 Only 7 years later, 10× Genomics released a data set of more than 1.3 million cells (https://support.10xgenomics.com/single-cell-gene-expression/datasets). The number of high impact studies has increased rapidly during the last few years, and in addition to transcriptional data, chromatin accessibility profiles can now be obtained at the single-cell level, enabling gene regulation studies and identification of cell type transcription factors and their targets.Citation3 ScRNA-Seq is able to detect and distinguish the transcriptome of individual cells; thus, the similarities and differences between cells can be studied. Previously unknown heterogeneity has been observed with distinct and rare cell populations being revealedCitation4,Citation5 in neuronsCitation6, embryos,Citation7,Citation8 and immune cells.Citation9,Citation10 Relationships between cell types can also be inferred allowing for insights into developmental progression and lineage tracing.Citation8,Citation11Citation15 Furthermore analysis of gene expression may reveal gene regulatory networks that control cell specificity, fate, and function.Citation16,Citation17

These rapidly emerging technologies also enable the detection and characterization of specific cell populations related to human development, health, and disease.Citation18 In parallel to the technological developments, multiple methods of bioinformatics analysis have been developed, including read quantification, quality control, normalization, clustering, cell annotation using reference maps, and identification of differentially expressed genes, cell surface markers, and ligand-receptor pairs. There has been rapid development of methods to reduce noise, improve sensitivity and, notably the throughput for single-cell transcriptomics.Citation19Citation23 To date, scRNA-Seq has been applied to mouse, primate, human fetal and adult retina,Citation24,Citation25 and to retinal organoids generated from pluripotent stem cells.Citation26Citation28 In this review, we take the opportunity to review this fast progress and summarize the current challenges and unanswered questions for the field of retinal development and disease.

scRNA-Seq technologies

There are a number of methods that have been developed for scRNA-Seq with differing approaches to cell capture, cDNA amplification, and library generation in order to isolate single cells and reduce biases.Citation5,Citation23,Citation29Citation38 The method used has an effect on the level of information obtained, such as the number of genes detected, number of transcripts per gene, expression of genes of interest, and resolution of differential splicing.Citation39 Despite this diversity in scRNA-Seq techniques, they all rely on the basic principles of viable single-cell preparation, isolation, lysis, capture of polyadenylated mRNA, reverse transcription, cDNA amplification, cDNA library preparation, and next-generation sequencing ().

Figure 1. Schematic presentation of single-cell capture, lysis, RNA/cDNA preparation and amplification, and library generation.

Figure 1. Schematic presentation of single-cell capture, lysis, RNA/cDNA preparation and amplification, and library generation.

Cells can be isolated in a number of ways. Tens to hundreds of cells can be captured in wells of a multi-well plate by fluorescence-activated cell sorting or smaller “capture sites” using microfluidics: this allows nanoliter volumes to be used, which can improve sensitivity.Citation40 Alternatively, droplet-based platforms such as Drop-SeqCitation37 and inDropCitation36 are able to capture thousands of cells in microdroplets, each containing a bead with a unique barcode and molecular identifier allowing generation of many scRNA-Seq libraries.Citation39 The number of cells and the methods employed for reverse transcription (RT), cDNA amplification, and sequencing library generation affects the ability to detect relative differences of transcript expression and reveal the biological variation between cells. However, as the amount of isolated RNA is relatively small from a single cell, scRNA-Seq is also prone to technical variability.Citation16,Citation21 This variation can occur during the conversion of a particular RNA into cDNA and its subsequent presence in the sequencing library, as a fraction of mRNAs is lost resulting in dropouts, or be affected by the fidelity of the amplification step introducing noise, or how accurately the mRNA input correlates to the number of reads.Citation41Citation43 The Smart-Seq method was developed to optimize the RT and amplification steps, and full-length transcript coverage. Further improvement led to Smart-Seq.2Citation32,Citation33 These methods are able to generate full-length cDNA libraries facilitating greater transcript coverage and detecting more genes per cell than alternative counting methods that employ unique identifying sequences.Citation43 A large portion of scRNA-Seq methods utilize these additional sequences in the RT primers. This reduces the transcript coverage but allows the inclusion of such sequences as barcodes, unique molecular identifiers, and adaptor sequences to identify a single RNA molecule and the cell of origin, as well as multiplexing and NGS library generation. The Unique Molecular Identifier (UMI) tags the mRNA at the RT stage prior to amplification allowing certain protocols such as CEL-Seq, inDrop, MARS-Seq, Drop-Seq, and SCRB-Seq to increase the throughput of library generation.Citation5,Citation23,Citation36Citation38 This can also distinguish cDNA and library amplification duplicatesCitation44 and improve the quantification of mRNA transcripts,Citation35,Citation42 which has been utilized in STRT-Seq,Citation35 CEL-Seq,Citation42 CEL-Seq2,Citation34 Drop-Seq,Citation37 inDrop,Citation36 MARS-Seq,Citation5 and SCRB-Seq.Citation38 Amplification-associated noise can also be reduced by in vitro transcription (IVT) rather than PCR amplification, which is utilized in CEL-Seq2 and MARS-Seq.Citation42,Citation43 Therefore, full-length Smart-Seq methods have fewer dropouts but greater amplification noise due to the use of PCR amplification. Methods utilizing IVT amplification (CEL-Seq2 and MARS-Seq) or UMIs (SCRB-Seq, CEL-Seq2, Drop-Seq, and MARS-Seq) have less amplification-associated noise.Citation42,Citation43 STRT-Seq enriches for the 5ʹ end of mRNA. CEL-Seq, CEL-Seq2, MARS-Seq, SCRB-Seq enrich for the 3ʹ end. All incorporate cell-specific barcodes and UMIs, facilitating pooling of cDNA for library generation, shortening the procedure. MARS-Seq increases the CEL-seq2 method throughput through the use of a liquid-handling platform.Citation5 If the aim is the quantification of transcriptomes from a large number of cells with a low sequencing depth then droplet-based approaches, e.g., Drop-Seq, are recommended. Whereas other methods such as SCRB-Seq and Smart-Seq2 are preferable for the quantification of fewer cells and greater sensitivity.Citation43

Miniaturization of the CEL-seq2 and Smart-Seq reactions to nanoliter volumes, as demonstrated by chip-based microfluidic systems, such as the Fluidigm platform, can improve sensitivity over standard scRNA-Seq.Citation45 The commercialization of these methods with proprietary hardware such as the Fluidigm C1 platform, as well as a number of droplet-based platforms, such as Chromium from 10x Genomics, ddSEQ from Bio-Rad Laboratories, InDrop from 1CellBio, and μEncapsulator from Dolomite Bio/Blacktrace Holdings is facilitating robust scRNA-Seq methodology for the masses. An alternative approach to scRNA-Seq is the isolation of single nuclei (sn) for snRNA-Seq. Studies have shown that despite the reduced number of transcripts from nuclei there is sufficient number to type them into broad classes of cells. Isolation of single nuclei may have some advantages over single cells as they are potentially less prone to any dissociation induced transcriptional changes and can be more easily isolated from complex and frozen tissues.Citation46Citation48

Computational methods and challenges

Single-cell RNA-Seq measures gene expression at the cellular level, meaning that distinct gene expression profiles of rare cell types are not masked by average expression. This gives the potential to answer questions that cannot be addressed using bulk RNA-Seq analysis. The analysis of such datasets can be used to identify cell populations using statistical clustering methods, to study changes from one developmental time point to another and pinpoint key regulatory genes.

Alignment and quantification

The analysis begins with the quantification of RNA by alignment of reads to a reference genome to create a gene by cell expression matrix. This process is very similar to bulk RNA-Seq analysis and many of the same tools are applicable to single-cell experiments. However, some specialized tools such as STARsolo which is an extension of the popular aligner STAR,Citation49 and Alevin, which is part of the Salmon toolkit are available for quantification of the reads detected. Additionally, a number of pipelines are available such as CellRanger,Citation50 which is distributed by 10x genomics for analysis of 10x datasets and DropEstCitation51 which can be used for the analysis of data from other platforms.

After the expression matrix has been created, the analysis methods start to deviate from bulk RNA-Seq analysis. Single-cell data are fundamentally different from bulk data and many of the assumptions made by statistical methods designed for bulk analysis do not hold true.Citation52 Single-cell data are sparse, with many genes either not detected or detected at very low levels; there are no replicates as each cell can only be measured once and the data is inherently noisy and prone to variation caused by technical artifacts. These qualities mean that a different analysis approach is required. Since 2015, the number of tools and analysis approaches has grown rapidly and there are now a rich array of methods, which can be applied to this data, the majority of which are written in R or Python. scRNA-toolsCitation53 is a comprehensive database cataloging the available methods. The omictools.org repository and ‘awesome-single-cell’Citation54 site also catalog a number of single-cell tools. The Bioconductor repositoryCitation55 hosts numerous packages for single-cell analysis as well as number of workflows explaining how these packages can be used in combination. In June 2019 Luecken and TheisCitation56 published a detailed article outlining the best practices for single-cell analysis. A general workflow for processing single-cell data is explained in . Briefly, the workflow consists of a pre-processing step involving quality control checks, normalization, and dimension reduction followed by statistical analysis to identify relationships between the single cells and key regulatory genes. These processes are described in more detail below.

Figure 2. Bioinformatics pipeline used for scRNA-Seq analysis.

Figure 2. Bioinformatics pipeline used for scRNA-Seq analysis.

Pre-processing – quality control

The analysis of the expression matrix begins with a quality control step. Dead cells or debris can obscure the interpretation of downstream analysis, so the gene expression must be filtered to remove problematic data. Poor quality data can be detected using metrics such as the number of genes detected per cell or the total counts per cell compared to the other cells in the experiment and filtering these cells from the analysis. The ScaterCitation57 and SeuratCitation58 R packages and the ScanPyCitation59 Python package provide tools and a framework for performing these initial quality control steps. Another source of technical noise comes in the form of doublets, where two or more cells are captured in the same bead or droplet. A number of tools now exist to predict doublets so that they can be filtered from the data.Citation60Citation63

Normalization and dimension reduction

Technical factors such as the number of genes detected per cell or the total number of reads per cell can contribute a significant proportion of cell-to-cell variability. Such factors must therefore be removed from the data by an appropriate normalization method before it is possible to measure the relationships between cells.Citation64,Citation65 scRNA-Seq data can be normalized by estimating a size factorCitation66 that is used to scale the data or using a probabilistic method to model the counts.Citation64,Citation67,Citation68

Where multiple samples are present in an analysis, a batch correction step may then be applied to the data. Batch correction methods designed for bulk samples can confound biological differences between cells with technical differences. A number of approaches have been designed for single-cell data. These include Mutual Nearest NeighborsCitation69 (MNN), Canonical Correlation AnalysisCitation70 (CCA), and Harmony.Citation71

After normalization and removal of batch effects, it is then necessary to reduce the dimensions of the dataset. This is because many of the genes measured show similar expression profiles across all cells so that despite the large number of data points captured for each cell, the distance between cells becomes very small, a phenomenon known as the “Curse of dimensionality”. To overcome this problem, the number of data points must be reduced so that only relevant sources of variation contribute to the interpretation of the data. This is achieved by a combination of feature selections where only the most biologically informative genes are used in the analysis and dimension reduction techniques. One method of feature selection that is commonly used is selecting highly variable genes. These are genes which show more variability than would be expected from the average expression of that gene.

Dimension reduction involves projecting the expression matrix into a low dimension space using techniques such as principal component analysis (PCA). These steps not only reduce the noise in the datasets but also help to reduce the computation time.Citation72

Cluster analysis

One of the key goals of scRNA-Seq is to identify cell types and subpopulations of cells within a dataset. This can be achieved using clustering analysis. Multiple tools exist for identifying clusters of cells in single-cell data, in fact there are currently 136 analysis tools listed in the scRNA-tools database. Some of the more popular packages include SeuratCitation58,Citation70 a graph-based method written in R, and Backspin – a biclusteringCitation73 method written in Python. Kiselev et alCitation72 published a comprehensive review with advice on selecting the most appropriate methods for single-cell clustering.

As many clustering methods involve estimating the number of clusters a priori or by using tuning parameters to set the resolution of the clusters, multiple clustering solutions can be found. Assessment and interpretation of the clusters require an understanding of the biology of the cells being studied. It is worth remembering that clusters can consist of true cell types (for example, Rods or Cones) or cell states, which reflect transitory functional differences between the cells. Clusters can be validated statisticallyCitation74 by measuring stability of clusters using methods such as silhouette analysisCitation75 or the elbow method. One useful tool for this task is ClustreeCitation76 which is designed to compare and evaluate multiple clustering solutions. Due to the complexity of scRNA-Seq datasets, it is useful to visualize in lower dimensional space – and the results of cluster analysis can be visualized using t-distributed stochastic neighbor embedding (t-SNE)Citation77 or Uniform Manifold Approximation and Projection for Dimension Reduction (UMAP).Citation78

Trajectory analysis

Clear groups of cells may not always be present within a sample. During development, cells may be transitioning from one state to another and it is therefore preferable to analyze this journey rather than perform clustering analysis, which forces partitions onto the data. Trajectory analysis can be used to study these continuous processes in cells. The analysis takes advantage of the asynchronous nature of cell development, and each cell is treated as a snapshot of the developmental pathway. The process begins by defining a path through the data and the cells are then ordered along this pathway. This creates a so-called pseudotime ordering to the cells in the experiment, allowing inferences to be made about the developmental trajectory of a process. More than 70 toolsCitation79 have already been developed for trajectory analysis capable of modeling linear, bifurcating or tree-shaped, or more complex topologies. Choosing the correct tool for a particular process is not an easy task. A comprehensive review of the available software was made by Saelens et al. in April. 2019Citation79 It concluded that the performance of a particular method was linked to the trajectory type being studied and that this should be used to guide the selection of the most appropriate method. For example, a linear trajectory may be sufficient when modeling the development of a single-cell type whereas a more complex tree-shaped trajectory may be necessary to analyze developmental processes of multiple cells.

Identification of differentially expressed genes

Differential expression analysis may be used to identify gene signatures for clusters. General statistical tests such as Wilcoxon rank-sum test as well as specialized methods, such as MAST,Citation68 ZINBWAVE,Citation80 and BaSICs,Citation81 which take into account zero inflation and high levels of technical noise specific to single-cell data can be applied, whereas for trajectory analysis a regression analysis or graph-autocorrelation analysis can be used to identify genes which change across a trajectory.

Cell type annotation and cell atlases

Whilst the above statistical methods provide insight into the level of heterogeneity and number of sub-populations of cells within a sample, to fully understand the data, the complex gene expression profiles must be annotated with known morphology or locations of cell types. This is a challenging process, which requires matching of marker genes detected by differential gene expression with known gene signatures for specific cell types, typically found through intensive literature searches. The annotation must then be validated by immunocytochemistry or other spatial methods.

In order to speed up this lengthy process, the Human Cell atlas project identified the need to create a detailed molecular map of every cell type in the human body, which could be used as a reference for future research. Cell atlases have the potential, when combined with cluster transfer tools such as scMapCitation82 and Garnet,Citation83 to dramatically speed up the cell annotation process.

Application of single-cell sequencing to studies of fetal and adult retina: cell type and marker identification

The retina is the innermost layer that lines the back of the eye and is vital for light sensing and image processing. The retina is derived from a germinal zone in the optic vesicle in which neuroepithelial cells proliferate to give rise to the six principle types of retinal neuronal cells (cones, rods, bipolar, amacrine, horizontal and ganglion cells) and one glial cell type (Müller glia), organized within three nuclear layers. All these cell types derive from retinal progenitor cells in an orderly spatio-temporal manner that has been well studied in vertebratesCitation84Citation86 and more recently in humans. Most of the retinal cells are sub-classified into subtypes; however, the molecular signature of each cell subtype remained unknown until the recent application of single-cell sequencing methods. The first study by Macosko et al. in 2015Citation87 used Dropseq to capture and analyze 44,808 cells from the retina of 14 old day mice, which identified 39 different cell clusters. This analysis was able to identify the known retinal cell types based on the specific expression of well-established retinal markers. Importantly, other cell types corresponding to astrocytes, microglia, endothelial cells, and fibroblasts were also identified and the relative abundance of each cell type was similar to previous estimates of microscopy–based techniques. Amacrine cells are the most diverse morphologically neuronal class being identified as inhibitory (utilizing GABA or glycine as neurotransmitter), excitatory (releasing glutamate) or amacrine cells with an unidentified neurotransmitter (nGnG amacrine cells). Macosko et al. were able to identify 21 clusters of amacrine cells in the adult mouse retina: of those, 12 were identified as GABAergic, 5 as glycinergic, 1 as excitatory, and 3 had low levels of GABAergic, glycinergic and glutamatergic markers, and hence these could represent the nGnG amacrine cells. Selective markers were then identified for each of the 21 amacrine cell subtypes, with the majority being further validated by immunohistochemistry.

Previous morphological and functional criteria have suggested the presence of around 100 cell types in the retina: this stands at odds with the above study, which identified only 39 cell types. This could be due to the number of cells captured and analyzed and the sequencing depth, which was perhaps insufficient to reveal diversity in the rare cell types (for example, retinal ganglion cells, which comprise <1% of retina). To overcome this, Rheaume et al. capturedCitation88 and analyzed 6225 retinal ganglion cells (RGCs) from the left- and right – eye mouse retina, using immuno-panning with the pan-RGC marker, Thy1. This analysis identified 40 RGC subtypes, corroborating a recent electrophysiological studyCitation89 and showed that all RGCs expressed the RBPMS marker, albeit at varying level. Most of the RGC subtypes were distributed proportionally between the left and the right eye; however, three clusters showed preferential distribution, indicating that some RGC related functions may be predominant in one eye. Using differential gene expression analysis, the study was able to identify subtype-specific markers, some of which were validated by fluorescence in situ hybridization (FISH). Interestingly the authors were also able to identify a new marker (Zic1) for the RGCs enriched in the right eye as well as transcription factor combinations for all the 40 RGC subtypes, which are documented in the RGC subtypes gene-browser: https://health.uconn.edu/neuroregenerationlab/rgc-subtypes-gene-browser.

Similarly, Shekhar et al.Citation90 used Dropseq to study mouse retinal bipolar cells (BCs) at single-cell resolution. BCs comprise around 7% of the retinal cells and are classified into rod and cone bipolar cells depending on the photoreceptor type from which they receive their synaptic input and further categorized into ON and OFF types based on the response to increases or decreases in light levels. Using a transgenic line that expresses GFP into All BCs and Müller Glia cells, the authors were able to capture and analyze around 25,000 cells from which 15 BC clusters could be identified, including all the know cell types and two novel populations, one of which displayed a non-typical BC morphology. The authors were also able to relate candidate marker expression to BC types using a combination of lentiviral labelling and FISH, which facilitated the assignment of new BC clusters, for example, BC1B with amacrine like morphology, which could have been incorrectly identified as an amacrine cell cluster, if only morphological-based classifications would have been applied.

Recently, Lukowski et al. were able to capture and analyze 20,009 cells from three adult human donor retinas and to identify 18 transcriptionally different cell populations.Citation91 The authors were able to show that there were no obvious variations in all major retinal cell types obtained from retina 6–14 hours post death with the exception of rod photoreceptors, which appeared to be more sensitive to degeneration. The single-cell sequencing data enabled the authors to identify a population with low MALAT1 expression, which correlated to degenerating rods. In an independent study, Voigt et al.Citation92 were able to capture 8,217 cells from three human adult neural retina obtained within 5.5 hours post death. Planned and reliable procurement of human adult retina is impossible; hence, authors cryopreserved the disassociated cells, which enabled stringent inclusion criteria and processing of samples obtained at different time points within one single cell-sequencing run. The authors were able to identify all the known retinal cell types, endothelial cells, and microglia within 17 clusters and to provide molecular markers, which should be of immense value for optimizing retinal organoid differentiation protocols as well as identification of novel candidate disease – genes. Both of these studies identified less cell clusters in human retina compared to the mouse. This is unlikely to be due to different cell type composition between human and mouse retina, but more likely to the deeper data mining within each cell cluster in the mouse retina, revealing cell type sub-specificity resulting in higher cluster number.

Voigt et al. were able to perform a regional difference expression analysis by characterizing 3578 cells from the fovea and 4639 cells from the peripheral human retina. The fovea is responsible for high acuity color vision and is characterized by a pit in which the inner retinal layers are displaced peripherally. Over the pit center, cone photoreceptor density is the highest in the retina, reaching over 200,000 cones/mm2. Rods are absent from the central 300um, which is called the rod free zone. Blood vessels are also absent forming the foveal avascular zone. A previous study has used bulk RNA-Seq analysis to characterize the development of the human fovea compared to the peripheral region;Citation85 nonetheless, Voight et al. provide the first regional single-cell analysis, identifying a total of 148 differentially expressed genes, 23 of which are enriched in foveal cells. Interestingly when the top 10 most enriched foveal and peripheral genes were compared to the single-cell study performed in M. fascularis,Citation93 85% similarity was observed, indicating the very high similarity in regional differences in retina transcriptome between humans and macaques. However, when the single-cell RNA-Seq data were compared between macaque and mouse retinal cell types, a tight correlation was found for photoreceptors, BCs, and amacrine cells; nonetheless, the RGCs were divergent between the two species, despite displaying conserved transcription factor codes. These findings are of great interest, and while suggesting a reliance on the mouse models for studies related to photoreceptor BC and amacrine cells function, they do question the validity of this model for investigating the function of genes related to RGC and function.

While the majority of the studies described above have focussed on adult human retina, a recent study published by Hu etCitation25 al has performed single-cell sequencing of 2421 cells from human fetal neural retina and the retinal pigment epithelium, encompassing the 5th until the 24th gestational week. Twenty-one clusters were identified comprising all the known retinal cell types as well as microglia, fibroblasts and blood cells. By analyzing peak marker expression, this study documented the emergence of RGCs and retinal progenitor cells (RPCs) at 5 weeks, followed by horizontal cells at 9 weeks and the later emergence of amacrine, photoreceptors, bipolar and Müller glia cells, corroborating bulk RNA-Seq studies of developing human retina.Citation85,Citation86 Importantly, the study identified cell-type specific transcription factors and their targets, which should facilitate immunohistochemical detection of these cell types both during in vivo and in vitro retinogenesis. Through mapping of the expression of known retinal disease genes to each cluster, the authors of this study were able to identify each gene to retinal-specific cells, thus shedding more light on the pathology of inherited retinal dystrophies.

Lineage tracing studies have shown that RPCs are multipotential, exhibit great variations in size, composition and division mode and can generate several if not all retinal cell types.Citation94Citation99 RPC fate is predominantly determined by the intrinsic cellular program with fine-tuning from extrinsic/environmental cues.Citation100Citation102 To better understand the transcriptional changes associated with changes in RPC competency, Clark et al.Citation103 performed a single-cell RNA-Seq of the developing mouse retina. Whilst this study identified a significant differential expression between RPCs at different stages, there were no sub-clusters within each individual age, indicating that RPCs undergo profound transcriptional changes during retinal development, which are consistent with the changes in their competency. The study identified embryonic day 16–18 as primary subdivision for early and late RPCs, with selective genes expressed in either of these two RPC subsets. The pseudotime analysis identified Nfi factors as being highly expressed within late-stage RPCs, bipolar and Müller glia cells. Accordingly, overexpression of NF1A/B/X resulted in an increase in the late-emerging retinal cells (BCs and Müller glia cells), while conditional loss of function alleles of Nfi genes in RPCs leads to a disruption of Müller glia marker staining and the outer limiting membrane. Together these new findings demonstrate that single-cell sequencing has the potential to identify new regulators of retinal histogenesis, in addition to cell type identification and generation of cell type markers ().

Figure 3. scRNA-Seq analysis of retina and pluripotent stem cell-derived retinal organoids: current insights.

Figure 3. scRNA-Seq analysis of retina and pluripotent stem cell-derived retinal organoids: current insights.

scRNA-Seq of pluripotent stem cell-derived retinal organoids: new insights into their cellular composition and cell fate determination

Three-dimensional (3D) structures combining multiple cell types, which interact with each other, named organoids have been used in classical developmental biology experiments since the early 1960s. These 3D organoids can be generated from pluripotent, adult and cancer stem cells and are widely used for studies of disease modeling, gene editing, drug testing/repurposing, and cell-based replacement therapies due to their ability to mimic human development and organogenesis.Citation104 To date, several protocols have been developed to optimize generation of various organoids including those of gut, kidney,Citation105,Citation106 liver,Citation107,Citation108 brain,Citation109 and retina. Traditionally the study of these organoids has relied on bulk transcriptomics and/or immunofluorescence characterization of various cell types based on known markers with the latter comprising a laborious and time-consuming technique, which is often difficult to implement on multiple time points and experimental groups. The power of scRNA-Seq to provide transcriptomic information of thousands of cells simultaneously has revolutionized the field of organoids and has started to generate new insights into their composition and cell maturationCitation110 as well as novel gene expression signatures as shown recently by Harder et al. and Czerniecki al., for human pluripotent stem cell-derived kidney organoids. Equally, scRNA-Seq has been used to compare 2D and 3D culture conditions for generation of liver bud-like structures from human pluripotent stem cells, showing that those generated under 3D culture conditions have a striking similarity to fetal liver cells.Citation107 Multiple studies have also applied scRNA-Seq to brain organoids and have provided evidence for the existence of progenitors and differentiated cells of both neuronal and mesenchymal lineages, resembling the fetal neocortex.Citation111 These studies have revealed that cerebral organoids have the ability to self-organize alongside the dorsal-ventral front and to give rise to mature astrocytes and oligodendrocytes in additional to neuronal lineages.Citation112 In some reported instances these brain organoids contain retina. Quadrato et al. performed scRNA-Seq on these photosensitive brain organoids and identified Müller glia, photoreceptors, retinal ganglion, bipolar, and amacrine cells of the neural retina as well as retinal pigment epithelium (RPE).Citation113

In a remarkable tour de force, Sasai and his group generated 3D aggregates from mouse embryonic stem cells, which under minimal culture condition went on to form optic vesicles and later optic cups displaying at later stages of differentiation process retinal stratification with typical apical basal polarity and correct spatial positioning of neural retina and RPE.Citation114 This process was soon replicated in human pluripotent stem cellsCitation115 with multiple groups using a combination of 2- and 3D protocols to generate human retinal organoids comprising all the retinal cell types within a laminated fashion that resembles the adult retina and responds to light.Citation116Citation119 Identification of each retinal cell type within these complex organoids is not easy as cell-type surface markers are far and few in between. However, gene-editing techniques have been used by Collin et al.,Citation120 and in 2017 Phillips et al.Citation121 to generate reporter cell lines harboring the green fluorescent protein (GFP) or td-Tomato at the Cone-Rod Homeobox (CRX) gene locus, a key transcriptional factor in retinal development. In both cases, scRNA-seq was performed allowing assessment of retinal organoid culture. Collin et al. enriched the CRX+ve precursors and used an unsupervised clustering approach, SC3,Citation122 which combines different clustering outcomes into a consensus matrix and displays each cell and differentially expressed genes within clusters. This resulted in the identification of 2 clusters with cluster 1 comprising 72% of the cells and showing gene expression akin to early cone photoreceptors and cluster 2 showing a transcriptional profile associated with an “early photoreceptor precursor”-like state. Importantly, when the authors transplanted the CRX+ve in an animal model of retinal degeneration, the transplanted cells showed a cone phenotype in vivo, corroborating the scRNA-Seq study and indicating the power of these technologies to predict cell fate determination prior to lengthy and costly in vivo studies. In the study performed by Phillips et al., the traditional unbiased principal component analysis (PCA) and hierarchical clustering methods were used but they did not reveal clearly and defined cell populations. Instead a new method, which generates graphical clusters based on the relative expression levels of bait genes, which ranks the single cells based on the level of expression that most closely matches that of the bait was used, bypassing the requirement for cell sorting and enrichment of photoreceptors prior to the application of scRNA-Seq.

The possibility to sequence higher cell numbers (1976 cells) allowed Collin et al.Citation27 to resolve the complexity of retinal organoids in a dataset combining day 60, 90 and 200 of differentiation, identifying 9 clusters and revealing the presence of precursor and mature cell types including cone and rod photoreceptors, Muller glia and RGCs as well as RPE cells. The rarer cell types such as horizontal, amacrine and bipolar cells were not identified despite being detectable by immunohistochemistry, suggesting that a higher cell number need to be sequenced in order to detect the rarer cell types within these complex organoids. A similar study was also performed by Kim et al.Citation123 in 8-month old cone-rich retinal organoids. Single-cell sequencing analysis indicated that 60% of the cells were photoreceptors with a ratio of cones to rods of 1.4:1. Moreover, this analysis demonstrated similarity between cones in the organoids and human macula, indicating the power of scRNA-Seq technologies to decipher the cell type, transcriptome, and maturity between in vitro and in vivo retinogenesis.

In addition to photoreceptor and full organoids, single-cell sequencing technologies have also been applied to the analysis of RGCs, the projection neurons of the retina that transmit the visual information to midbrain through the optic nerve. Daniszewki et al.,Citation124 analysed 1,714 embryonic stem cell-derived RGC (enriched on the basis of reporter expression (Brn3b-mCherry)) and identified 3 distinct subpopulations of cells that included progenitors as well as mature RGCs, which contained genes related to the axon guidance together with the semaphoring interactions, extracellular matrix proteins and a downregulation of cell cycle genes. In a similar study, Langer et al.Citation125 were able to distinguish several RGC subtypes as well as molecular markers, which could be invaluable for improving and assessing the efficiency of differentiation protocols. It is known that each RGC subtype plays a different role in the visual pathway; in fact, molecular markers remain unknown for primates, including humans.Citation126 Langer et al. identified DCX as a new candidate gene for the direction-selective RGCs (DS-RGCs) and provided new insights on the abundance of each RGC subtype based on expression of RGC specific subtype markers. Taken together all these data point towards the utility of scRNA-Seq in deciphering the cell composition of pluripotent stem cell retinal organoids and retinal cells, discovering novel subtype-specific cell markers and assessing their similarity and maturation state in comparison to the equivalent cell types in the developing and adult retina. As the scRNA-Seq techniques improve and costs get lower, their application to drug repurposing/discovery and disease-modeling studies becomes more feasible and tangible with potential to deliver important insights for patients with retinal disease in a short time frame.

Conclusions and final remarks

Since the early work of Ramon y Cajal, depicting and drawing the various types of retinal cells and their interactions,Citation127 a lot of thought and effort has gone towards the retinal cellular classification by a combination of approaches including light and transmission electron microscopy, molecular biology, FISH, optical coherence tomography, etc. While distinctions at the higher levels of the cellular hierarchy, based on morphology, physiology, and gene expression, are relatively straightforward to make, it is more difficult to achieve the finer divisions of cellular subtypes with precision. This is where single-cell sequencing methods are making a huge and rapidly growing contribution to cell type specification of many human organs from development to adulthood as well generation of cell-type specific markers as summarized in this current review. Despite this remarkable progress, there remain unresolved questions. For example, how similar/different do the transcriptional profiles of the cells have to be in order to be associated/segregated into the same/different subtype? Are there profound differences in cell subtypes from the left and the right retina? How do we match the cell number and subtypes identified by different methods? How many methods are needed to validate the results of single-cell sequencing? From how many donors does one need to perform single-cell capture and analysis to achieve consistent and reliable results? What is the ideal cell number and sequencing depth for each tissue to achieve the best cellular and subcellular classification? How does single cell disassociation and time from retrieval affect the viability of different retinal cell types and the single-cell data? To date, the handful of studies performed in mouse, primate and human retina, as discussed in the main body of the review, have not managed to dissect all 100-cell types shown by microscopy and electrophysiological findings. This indicates that the cell number and sequencing depth reported in these initial studies are probably insufficient to reveal the complexity of adult retina. The success in classifying BCsCitation90 with shallow sequencing suggests that capturing more cells and sequencing at a lower read depth can be used for comprehensive cellular classification. This can be followed by resequencing of particular subsets, which are more dynamic and complex in their transcriptional profile, at a higher depth. In addition, advances made in the field of single-cell sequencing and continuously decreasing sequencing costs will permit single-cell studies in many labs with increased cell numbers. Combined analysis of datasets from multiple donors performed in different laboratories with appropriate computational methods that correct for batch sequencing effects, coupled with extensive validations, could provide a solid foundation for generating the cellular hierarchy and retinal cell-specific markers both during development and in adulthood.

Exciting developments in the field of spatial transcriptomicsCitation128 and light sheet microscopyCitation129 have started to provide the necessary high spatiotemporal resolution. The complexity of these methods, the skills and equipment and the specificity of each tissue response to application of such techniques make it difficult to envisage how they could be commonplace practice in every laboratory. However, as seen with scRNA-Seq, rapid advancement, optimization, and commercialization (e.g., 10x Genomics) will lead to wider adoption of such techniques. Furthermore, strategic initiatives such as the Human Cell Atlas (HCA), which bring together excellent groups of scientists and bioinformaticians, will provide the technological and data-driven online accessible resource for all those interested in further studies. These could be directed at confirmation of gene expression at protein level and how this changes during development; others could compare the developmental and adult single-cell expression data to retinal organoids grown in their own laboratories to stage and assess the developmental maturity and cell type needed for transplantation or toxicology-based studies. Importantly, the wealth of data provided by single-cell sequencing together with regional and spatial maps provides an excellent resource for mapping the expression of known disease genes to certain cell types, which will facilitate better disease understanding as well as therapeutic interventions. Finally, yet importantly, identification of novel regulators during retinal development is going to provide the necessary baseline for identifying new disease genes, which has not been possible to date due to mutations in non-coding sequences or lack of understanding about the expression of the disease gene in particular retinal cell types. scRNA-Seq coupled with single-cell chromatin accessibility (for example ATAC-Seq) and bioinformatics analyses (pseudotime, etc.) are starting to identify the gene regulatory networks and their dynamics across species, cell surface markers and ligand-receptor pairs at a scale that was probably unimaginable during the previous decade. The question now is not when and at what scale, but how biologists, clinicians, and lawmakers, will best use this information to help treat patients with inherited and age-related retinal disease. Perhaps the experience set by the UK in exploiting the data generated from the 100,000 genomes could provide an enlightening example of how technological revolutions can bring about improvement in healthcare and life quality of many patients worldwide and bring the bench research closer to the bedside.

Additional information

Funding

We are grateful to Medical Research Council UK [#MR/S035826/1 and #MR/S036237/1] for funding our current work on single cell sequencing in the developing and adult eye and ERC [#614620] for funding our work on single cell sequencing of retinal organoids.

References

  • Wang YJ, Kaestner KH. Single-cell RNA-seq of the pancreatic islets––a promise not yet fulfilled? Cell Metab. [Internet]. 2019 Mar 5;29(3):539–44. [accessed 2019 Aug 13]. http://www.ncbi.nlm.nih.gov/pubmed/30581120
  • Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, Wang X, Bodeau J, Tuch BB, Siddiqui A, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. [Internet]. 2009 May 6;6(5):377–82. accessed 2019 Aug 13. http://www.nature.com/articles/nmeth.1315
  • Clark SJ, Lee HJ, Smallwood SA, Kelsey G, Reik W. Single-cell epigenomics: powerful new methods for understanding gene regulation and cell identity. Genome Biol. 2016 Apr 18;17(1):72.
  • Deng Q, Ramsköld D, Reinius B, Sandberg R. Single-cell RNA-seq reveals dynamic, random monoallelic gene expression in mammalian cells. Science (80-). 2014;343:193–96.
  • Jaitin DA, Kenigsberg E, Keren-Shaul H, Elefant N, Paul F, Zaretsky I, Mildner A, Cohen N, Jung S, Tanay A, et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science (80-). 2014;343(6172):776–79.
  • Zeisel A, M͡oz-Manchado AB, Codeluppi S, Lönnerberg P, Manno GL, Juréus A, Marques S, Munguba H, He L, Betsholtz C, et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science (80-). 2015 Mar 6;347(6226):1138–42.
  • Yan L, Yang M, Guo H, Yang L, Wu J, Li R, Liu P, Lian Y, Zheng X, Yan J, et al. Single-cell RNA-Seq profiling of human preimplantation embryos and embryonic stem cells. Nat Struct Mol Biol. 2013 Sep;20(9):1131–39.
  • Blakeley P, Fogarty NME, Del Valle I, Wamaitha SE, Hu TX, Elder K, Snell P, Christie L, Robson P, Niakan KK. Defining the three cell lineages of the human blastocyst by single-cell RNA-seq. Dev. 2015 Sep 23;142(18):3151–65.
  • Shalek AK, Satija R, Shuga J, Trombetta JJ, Gennert D, Lu D, Chen P, Gertner RS, Gaublomme JT, Yosef N, et al. Single-cell RNA-seq reveals dynamic paracrine control of cellular variation. Nature. 2014 Jun;510(7505):363–69.
  • Stubbington MJT, Lönnberg T, Proserpio V, Clare S, Speak AO, Dougan G, Teichmann SA. T cell fate and clonality inference from single-cell transcriptomes. Nat Methods. 2016 Apr;13(4):329–32.
  • Treutlein B, Brownfield DG, Wu AR, Neff NF, Mantalas GL, Espinoza FH, Desai TJ, Krasnow MA, Quake SR. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature. 2014 May;509(7500):371–75.
  • Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, Livak KJ, Mikkelsen TS, Rinn JL. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014 Apr;32(4):381–86.
  • Petropoulos S, Edsgärd D, Reinius B, Deng Q, Panula SP, Codeluppi S, Plaza Reyes A, Linnarsson S, Sandberg R, Lanner F. Single-cell RNA-seq reveals lineage and x chromosome dynamics in human preimplantation embryos. Cell. 2016 May;165(4):1012–26.
  • Lönnberg T, Svensson V, James KR, Fernandez-Ruiz D, Sebina I, Montandon R, Soon MSF, Fogg LG, Nair AS, Liligeto UN, et al. Single-cell RNA-seq and computational analysis using temporal mixture modeling resolves TH1/TFH fate bifurcation in malaria. Sci Immunol. 2017 Mar;2(9):eaal2192.
  • Venteicher AS, Tirosh I, Hebert C, Yizhak K, Neftel C, Filbin MG, Hovestadt V, Escalante LE, Shaw ML, Rodman C, et al. Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science. [Internet]. 2017;355(6332):eaai8478. accessed 2019 Sep 2. http://www.ncbi.nlm.nih.gov/pubmed/28360267
  • Wagner A, Regev A, Yosef N. Revealing the vectors of cellular identity with single-cell genomics. Nat Biotechnol. 2016 Nov;34(11):1145–60.
  • Liu S, Trapnell C. Single-cell transcriptome sequencing: recent advances and remaining challenges. F1000Res. 2016 Feb 17;5. pii: F1000 Faculty Rev-182. doi:10.12688/f1000research.7223.1eCollection 2016.
  • Young MD, Mitchell TJ, Vieira Braga FA, Tran MGB, Stewart BJ, Ferdinand JR, Collord G, Botting RA, Popescu D-M, Loudon KW, et al. Single-cell transcriptomes from human kidneys reveal the cellular identity of renal tumors. Science. [Internet]. 2018;361(6402):594–99. [accessed 2019 Sep 2]. http://www.ncbi.nlm.nih.gov/pubmed/30093597
  • Wilson NK, Kent DG, Buettner F, Shehata M, Macaulay IC, Calero-Nieto FJ, Sánchez Castillo M, Oedekoven CA, Diamanti E, Schulte R, et al. Combined single-cell functional and gene expression analysis resolves heterogeneity within stem cell populations. Cell Stem Cell. [Internet]. 2015 Jun 4;16(6):712–24. [accessed 2019 Aug 13]. http://www.ncbi.nlm.nih.gov/pubmed/26004780
  • Islam S, Zeisel A, Joost S, La Manno G, Zajac P, Kasper M, Lönnerberg P, Linnarsson S. Quantitative single-cell RNA-seq with unique molecular identifiers. Nat Methods. [Internet]. 2014 Feb 22;11(2):163–66. [accessed 2019 Aug 13]. http://www.nature.com/articles/nmeth.2772
  • Kolodziejczyk AA, Kim JK, Svensson V, Marioni JC, Teichmann SA. The technology and biology of single-cell RNA sequencing. Mol Cell. [Internet]. 2015 May 21;58(4):610–20. [accessed 2019 Aug 13]. https://linkinghub.elsevier.com/retrieve/pii/S1097276515002610
  • Grün D, van Oudenaarden A. Design and analysis of single-cell sequencing experiments. Cell. [Internet]. 2015 Nov 5;163(4):799–810. [accessed 2019 Aug 13]. https://linkinghub.elsevier.com/retrieve/pii/S0092867415013537
  • Hashimshony T, Wagner F, Sher N, Yanai I. CEL-Seq: single-cell RNA-Seq by multiplexed linear amplification. Cell Rep. [Internet]. 2012 Sep 27;2(3):666–73. [accessed 2019 Aug 13]. https://linkinghub.elsevier.com/retrieve/pii/S2211124712002288
  • Welby E, Lakowski J, Di Foggia V, Budinger D, Gonzalez-Cordero A, Lun ATL, Epstein M, Patel A, Cuevas E, Kruczek K, et al. Isolation and comparative transcriptome analysis of human fetal and iPSC-derived cone photoreceptor cells. Stem Cell Rep. [Internet]. 2017 Dec 12;9(6):1898–915. [accessed 2018 Mar 1]. http://www.ncbi.nlm.nih.gov/pubmed/29153988
  • Hu Y, Wang X, Hu B, Mao Y, Chen Y, Yan L, Yong J, Dong J, Wei Y, Wang W, et al. Dissecting the transcriptome landscape of the human fetal neural retina and retinal pigment epithelium by single-cell RNA-seq analysis. Demb J, editor. PLoS Biol. [Internet]. 2019 Jul 3;17(7):e3000365. [accessed 2019 Aug 14]. http://www.ncbi.nlm.nih.gov/pubmed/31269016
  • Phillips MJ, Jiang P, Howden S, Barney P, Min J, York NW, Chu LF, Capowski EE, Cash A, Jain S, et al. A novel approach to single cell RNA-sequence analysis facilitates in silico gene reporting of human pluripotent stem cell-derived retinal cell types. Stem Cells. 2018 Mar;36(3):313–24.
  • Collin J, Queen R, Zerti D, Dorgau B, Hussain R, Coxhead J, Cockell S, Lako M. Deconstructing retinal organoids: single cell RNA-seq reveals the cellular components of human pluripotent stem cell-derived retina. Stem Cells. [Internet]. 2019 Jan 12. [accessed 2019 Mar 14]. http://www.ncbi.nlm.nih.gov/pubmed/30548510
  • Collin J, Zerti D, Queen R, Santos-Ferreira T, Bauer R, Coxhead J, Hussain R, Steel D, Mellough C, Ader M, et al. CRX expression in pluripotent stem cell-derived photoreceptors marks a transplantable subpopulation of early cones. Stem Cells. [Internet]. 2019 Jan 30; cited 2019 Mar 14. http://www.ncbi.nlm.nih.gov/pubmed/30681766
  • Velten L, Anders S, Pekowska A, Järvelin AI, Huber W, Pelechano V, Steinmetz LM. Single‐cell polyadenylation site mapping reveals 3′ isoform choice variability. Mol Syst Biol. 2015 Jun;11(6):812.
  • Islam S, Kjällquist U, Moliner A, Zajac P, Fan J-B, Lönnerberg P, Linnarsson S. Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq.Genome Res. [Internet]. 2011 Jul;21(7):1160–67. [accessed 2019 Sep 2]. http://www.ncbi.nlm.nih.gov/pubmed/21543516
  • Verboom K, Everaert C, Bolduc N, Livak KJ, Yigit N, Rombaut D, Anckaert J, Lee S, Venø MT, Kjems J, et al. SMARTer single cell total RNA sequencing. Nucleic Acids Res. 2019 Sep 19;47(16):e93.
  • Picelli S, Björklund ÅK, Faridani OR, Sagasser S, Winberg G, Sandberg R. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat Methods. 2013 Nov;10(11):1096–98.
  • Picelli S, Faridani OR, Björklund ÅK, Winberg G, Sagasser S, Sandberg R. Full-length RNA-seq from single cells using smart-seq2. Nat Protoc. 2014 Jan;9(1):171–81.
  • Hashimshony T, Senderovich N, Avital G, Klochendler A, de Leeuw Y, Anavy L, Gennert D, Li S, Livak KJ, Rozenblatt-Rosen O, et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. [Internet]. 2016 Apr 28;17:77. accessed 2019 Sep 2. http://www.ncbi.nlm.nih.gov/pubmed/27121950
  • Islam S, Zeisel A, Joost S, La Manno G, Zajac P, Kasper M, Lönnerberg P, Linnarsson S. Quantitative single-cell RNA-seq with unique molecular identifiers. Nat Methods. 2014 Feb;11(2):163–66.
  • Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L, Weitz DA, Kirschner MW. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell. 2015 May 30;161(5):1187–201.
  • Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 2015 May;161(5):1202–14.
  • Soumillon M, Cacchiarelli D, Semrau S, Oudenaarden AV, Mikkelsen TS. Characterization of directed differentiation by high-throughput single-cell RNA-Seq. bioRxiv. 2014 Mar:003236.
  • Haque A, Engel J, Teichmann SA, Lönnberg T. A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med. 2017 Aug 18;9(1):75. doi:10.1186/s13073-017-0467-4
  • Svensson V, Natarajan KN, Ly L-H, Miragaia RJ, Labalette C, Macaulay IC, Cvejic A, Teichmann SA. Power analysis of single-cell RNA-sequencing experiments. Nat Methods. 2017 Apr;14(4):381–87.
  • Brennecke P, Anders S, Kim JK, Kołodziejczyk AA, Zhang X, Proserpio V, Baying B, Benes V, Teichmann SA, Marioni JC, et al. Accounting for technical noise in single-cell RNA-seq experiments. Nat Methods. 2013 Nov;10(11):1093–95.
  • Grün D, Kester L, van Oudenaarden A. Validation of noise models for single-cell transcriptomics. Nat Methods. 2014 Jun;11(6):637–40.
  • Ziegenhain C, Vieth B, Parekh S, Reinius B, Guillaumet-Adkins A, Smets M, Leonhardt H, Heyn H, Hellmann I, Enard W. Comparative analysis of single-cell RNA sequencing methods. Mol Cell. 2017 Feb 16;65(4):631–643.e4.
  • Kivioja T, Vähärautio A, Karlsson K, Bonke M, Enge M, Linnarsson S, Taipale J. Counting absolute numbers of molecules using unique molecular identifiers. Nat Methods. 2012 Jan;9(1):72–74.
  • Wu AR, Neff NF, Kalisky T, Dalerba P, Treutlein B, Rothenberg ME, Mburu FM, Mantalas GL, Sim S, Clarke MF, et al. Quantitative assessment of single-cell RNA-sequencing methods. Nat Methods. 2014 Jan;11(1):41–46.
  • Bakken TE, Hodge RD, Miller JA, Yao Z, Nguyen TN, Aevermann B, Barkan E, Bertagnolli D, Casper T, Dee N, et al. Single-nucleus and single-cell transcriptomes compared in matched cortical cell types. PLoS One. 2018 Dec 1;13(12):e0209648.
  • Lake BB, Codeluppi S, Yung YC, Gao D, Chun J, Kharchenko PV, Linnarsson S, Zhang K. A comparative strategy for single-nucleus and single-cell transcriptomes confirms accuracy in predicted cell-type expression from nuclear RNA. Sci Rep. 2017 Dec 1;7(1):6031.
  • Grindberg RV, Yee-Greenbaum JL, McConnell MJ, Novotny M, O’Shaughnessy AL, Lambert GM, Araúzo-Bravo MJ, Lee J, Fishman M, Robbins GE, et al. RNA-sequencing from single nuclei. Proc Natl Acad Sci. [Internet]. 2013 Dec 3;110(49):19802–07. accessed 2019 Oct 22. https://www.pnas.org/content/110/49/19802
  • Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. [Internet]. 2012;29(1):15–21. doi:10.1093/bioinformatics/bts635.
  • Zheng GXY, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, Ziraldo SB, Wheeler TD, McDermott GP, Zhu J, et al. Massively parallel digital transcriptional profiling of single cells. Nat Commun. [Internet]. 2017;8:14049. doi:10.1038/ncomms14049.
  • Petukhov V, Guo J, Baryawno N, Severe N, Scadden DT, Samsonova MG, Kharchenko PV. DropEst: pipeline for accurate estimation of molecular counts in droplet-based single-cell RNA-seq experiments. Genome Biol. [Internet]. 2018;19(1):78. doi:10.1186/s13059-018-1449-6.
  • Stegle O, Teichmann SA, Marioni JC. Computational and analytical challenges in single-cell transcriptomics. Nat Rev Genet. [Internet]. 2015;16:133. doi:10.1038/nrg3833.
  • Zappia L, Phipson B, Oshlack A. Exploring the single-cell RNA-seq analysis landscape with the scRNA-tools database. PLoS Comput Biol. [Internet]. 2018;14(6):e1006245. doi:10.1371/journal.pcbi.1006245.
  • Davis S, Kutum R, Zappia L, Sorenson J, Kiselev V, olivier P, Botvinnik O, Korthauer K, Gitter A, Campbell KR, et al. seandavi/awesome-single-cell: 2018-06-20-1. 2018 Jun 20.
  • Amezquita RA, Carey VJ, Carpp LN, Geistlinger L, Lun ATL, Marini F, Rue-Albrecht K, Risso D, Soneson C, Waldron L, et al. Orchestrating single-cell analysis with bioconductor. bioRxiv. [Internet]. 2019. 590562. https://www.biorxiv.org/content/biorxiv/early/2019/03/27/590562.full.pdf
  • Luecken MD, Theis FJ. Current best practices in single-cell RNA-seq analysis: a tutorial. Mol Syst Biol. [Internet]. 2019;15(6):e8746–e8746. https://www.ncbi.nlm.nih.gov/pubmed/31217225
  • McCarthy DJ, Campbell KR, Lun ATL, Wills QF. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in. R Bioinformatics. [Internet]. 2017;33(8):1179–86. doi:10.1093/bioinformatics/btw777.
  • Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck III WM, Hao Y, Stoeckius M, Smibert P, Satija R. Comprehensive integration of single-cell data. Cell. [Internet]. 2019;177(7):1888–1902.e21. doi:10.1016/j.cell.2019.05.031.
  • Wolf FA, Angerer P, Theis FJ. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. [Internet]. 2018;19(1):15. doi:10.1186/s13059-017-1382-0.
  • DePasquale EAK, Schnell DJ, Valiente-Alandí Í, Blaxall BC, Grimes HL, Singh H, Salomonis N. DoubletDecon: cell-state aware removal of single-cell RNA-seq doublets. bioRxiv. [Internet]. 2018:364810. https://www.biorxiv.org/content/biorxiv/early/2018/07/08/364810.full.pdf
  • McGinnis CS, Murrow LM, Gartner ZJ. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. [Internet]. 2019;8(4):329–337.e4. http://www.sciencedirect.com/science/article/pii/S2405471219300730
  • Bais AS, Kostka D. scds: computational annotation of doublets in single cell RNA sequencing data. bioRxiv. [Internet]. 2019:564021. https://www.biorxiv.org/content/biorxiv/early/2019/02/28/564021.full.pdf
  • Wolock SL, Lopez R, Klein AM. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. [Internet]. 2019;8(4):281–291.e9. doi:10.1016/j.cels.2018.11.005.
  • Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. bioRxiv. [Internet]. 2019:576827. http://biorxiv.org/content/early/2019/03/18/576827.abstract
  • Cole MB, Risso D, Wagner A, DeTomaso D, Ngai J, Purdom E, Dudoit S, Yosef N. Performance assessment and selection of normalization procedures for single-cell RNA-seq. Cell Syst. 2019 Apr 24;8(4):315–328.e8.
  • Lun ATL, Bach K, Marioni JC. Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome Biol. [Internet]. 2016;17:75. https://www.ncbi.nlm.nih.gov/pubmed/27122128
  • Vallejos CA, Risso D, Scialdone A, Dudoit S, Marioni JC. Normalizing single-cell RNA sequencing data: challenges and opportunities. Nat Methods. [Internet]. 2017 May 15;14(6):565–71. https://www.ncbi.nlm.nih.gov/pubmed/28504683.
  • Finak G, McDavid A, Yajima M, Deng J, Gersuk V, Shalek AK, Slichter CK, Miller HW, McElrath MJ, Prlic M, et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. [Internet]. 2015;16:278. https://www.ncbi.nlm.nih.gov/pubmed/26653891
  • Haghverdi L, Lun ATL, Morgan MD, Marioni JC. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat Biotechnol. [Internet]. 2018 Apr 02;36(5):421–27. https://www.ncbi.nlm.nih.gov/pubmed/29608177.
  • Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. [Internet]. 2018;36:411. doi:10.1038/nbt.4096.
  • Korsunsky I, Fan J, Slowikowski K, Zhang F, Wei K, Baglaenko Y, Brenner M, Loh P-R, Raychaudhuri S. Fast, sensitive, and accurate integration of single cell data with Harmony. bioRxiv. [Internet]. 2018:461954. https://www.biorxiv.org/content/biorxiv/early/2018/11/05/461954.full.pdf
  • Kiselev VY, Andrews TS, Hemberg M. Challenges in unsupervised clustering of single-cell RNA-seq data. Nat Rev Genet. [Internet]. 2019;20(5):273–82. doi:10.1038/s41576-018-0088-9.
  • Zeisel A, Muñoz-Manchado AB, Codeluppi S, Lönnerberg P, La Manno G, Juréus A, Marques S, Munguba H, He L, Betsholtz C, et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science (80-) [Internet]. 2015;347:1138–42. https://science.sciencemag.org/content/sci/347/6226/1138.full.pdf
  • Ketchen DJ, Shook CL. The application of cluster analysis in strategic management research: an analysis and critique. Strateg Manag J. 1996;17:441–58.
  • Lovmar L, Ahlford A, Jonsson M, Syvänen AC. Silhouette scores for assessment of SNP genotype clusters. BMC Genomics. 2005 Mar 10;6(1):35.
  • Zappia L, Oshlack A. Clustering trees: a visualization for evaluating clusterings at multiple resolutions. Gigascience. [Internet]. 2018;7(7):giy083. doi:10.1093/gigascience/giy083.
  • van der Maaten L, Hinton G. Visualizing data using t-SNE. J Mach Learn Res. 2008;9:2579–605.
  • McInnes L, Healy J, Melville J UMAP: uniform manifold approximation and projection for dimension reduction. 2018 Feb 9 [accessed 2019 Sep 2]. http://arxiv.org/abs/1802.03426
  • Saelens W, Cannoodt R, Todorov H, Saeys Y. A comparison of single-cell trajectory inference methods. Nat Biotechnol. [Internet]. 2019;37(5):547–54. doi:10.1038/s41587-019-0071-9.
  • Risso D, Perraudeau F, Gribkova S, Dudoit S, Vert J-P. A general and flexible method for signal extraction from single-cell RNA-seq data. Nat Commun. [Internet]. 2018;9(1):284. doi:10.1038/s41467-017-02554-5.
  • Vallejos CA, Marioni JC, Richardson S. BASiCS: bayesian analysis of single-cell sequencing data. PLoS Comput Biol. [Internet]. 2015;11(6):e1004333. doi:10.1371/journal.pcbi.1004333.
  • Kiselev VY, Yiu A, Hemberg M. scmap: projection of single-cell RNA-seq data across data sets. Nat Methods. [Internet]. 2018;15:359. doi:10.1038/nmeth.4644.
  • Pliner HA, Shendure J, Trapnell C. Supervised classification enables rapid annotation of cell atlases. bioRxiv. [Internet]. 2019:538652. https://www.biorxiv.org/content/biorxiv/early/2019/02/04/538652.full.pdf
  • Marquardt T, Gruss P. Generating neuronal diversity in the retina: one for nearly all.Trends Neurosci. [Internet]. 2002 Jan;25(1):32–38. [accessed 2019 Sep 2]. http://www.ncbi.nlm.nih.gov/pubmed/11801336
  • Hoshino A, Ratnapriya R, Brooks MJ, Chaitankar V, Wilken MS, Zhang C, Starostik MR, Gieser L, La Torre A, Nishio M, et al. Molecular anatomy of the developing human retina. Development. 2019 Jan 29;142(2):pii: dev169474. doi:10.1242/dev.169474
  • Mellough CB, Roman B, Joseph C, Dorgau B, Zerti D, Dolan DWP, Jones CM, Izuogu OG, Min Y, Dean H, et al. An integrated transcriptional analysis of the developing human retina. Under Rev. 2018;146(2):dev169474.
  • Macosko EZ, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, Trombetta JJ, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 2015;161:1202–14.
  • Rheaume BA, Jereen A, Bolisetty M, Sajid MS, Yang Y, Renna K, Sun L, Robson P, Trakhtenberg EF. Single cell transcriptome profiling of retinal ganglion cells identifies cellular subtypes. Nat Commun. [Internet]. 2018;9(1):2759. [accessed 2019 Sep 2]. http://www.ncbi.nlm.nih.gov/pubmed/30018341
  • Baden T, Berens P, Franke K, Román Rosón M, Bethge M, Euler T. The functional diversity of retinal ganglion cells in the mouse. Nature. 2016 Jan 21;529(7586):345–50.
  • Shekhar K, Lapan SW, Whitney IE, Tran NM, Macosko EZ, Kowalczyk M, Adiconis X, Levin JZ, Nemesh J, Goldman M, et al. Comprehensive classification of retinal bipolar neurons by single-cell transcriptomics. Cell. 2016 Aug 25;166(5):1308–1323.e30.
  • Lukowski SW, Lo CY, Sharov AA, Nguyen Q, Fang L, Hung SS, Zhu L, Zhang T, Grünert U, Nguyen T, et al. A single‐cell transcriptome atlas of the adult human retina. Embo J. [Internet]. 2019 Aug 22;38(18). [accessed 2019 Sep 2]. https://doi.org/10.15252/embj.2018100811
  • Voigt AP, Whitmore SS, Flamme-Wiese MJ, Riker MJ, Wiley LA, Tucker BA, Stone EM, Mullins RF, Scheetz TE. Molecular characterization of foveal versus peripheral human retina by single-cell RNA sequencing. Exp Eye Res. 2019 Jul 1;184:234–42.
  • Peng YR, Shekhar K, Yan W, Herrmann D, Sappington A, Bryman GS, van Zyl T, Do MTH, Regev A, Sanes JR. Molecular classification and comparative taxonomics of foveal and peripheral cells in primate retina. Cell. 2019 Feb 21;176(5):1222–1237.e22.
  • Cepko CL, Austin CP, Yang X, Alexiades M, Ezzeddine D. Cell fate determination in the vertebrate retina. Proc Natl Acad Sci. [Internet]. 1996 Jan 23;93(2):589–95. [accessed 2019 Sep 2]. https://doi.org/10.1073/pnas.93.2.589
  • Livesey FJ, Cepko CL. Vertebrate neural cell-fate determination: lessons from the retina. Nat Rev Neurosci. 2001;2:109–18.
  • Turner DL, Cepko CL. A common progenitor for neurons and glia persists in rat retina late in development.Nature. [Internet]. 1987 Jul;328(6126):131–36. accessed 2019 Sep 2. http://www.nature.com/articles/328131a0
  • Cayouette M, Barres BA, Raff M. Importance of intrinsic mechanisms in cell fate decisions in the developing rat retina. Neuron. 2003 Dec 4;40(5):897–904.
  • Rapaport DH, Patheal SL, Harris WA. Cellular competence plays a role in photoreceptor differentiation in the developing Xenopus retina. J Neurobiol. [Internet]. 2001 Nov 5;49(2):129–41. [accessed 2019 Sep 2]. http://www.ncbi.nlm.nih.gov/pubmed/11598920
  • Belliveau MJ, Cepko CL. Extrinsic and intrinsic factors control the genesis of amacrine and cone cells in the rat retina.Development. [Internet]. 1999 Feb;126(3):555–66. accessed 2019 Sep 2. http://www.ncbi.nlm.nih.gov/pubmed/9876184
  • Xiang M. Intrinsic control of mammalian retinogenesis. Cell Mol Life Sci. 2013;70:2519–32.
  • Bassett EA, Wallace VA. Cell fate determination in the vertebrate retina. Trends Neurosci. 2012;35:565–73.
  • Gomes FLAF, Zhang G, Carbonell F, Correa JA, Harris WA, Simons BD, Cayouette M. Reconstruction of rat retinal progenitor cell lineages in vitro reveals a surprising degree of stochasticity in cell fate decisions. Development. 2011 Jan 15;138(2):227–35.
  • Clark BS, Stein-O’Brien GL, Shiau F, Cannon GH, Davis-Marcisak E, Sherman T, Santiago CP, Hoang TV, Rajaii F, James-Esposito RE, et al. Single-cell RNA-seq analysis of retinal development identifies NFI factors as regulating mitotic exit and late-born cell specification. Neuron. 2019 Jun 19;102(6):1111–1126.e5.
  • Kretzschmar K, Clevers H. Organoids: modeling development and the stem cell niche in a dish. Dev Cell. [Internet]. 2016;38(6):590–600. [accessed 2019 Aug 27]. http://www.ncbi.nlm.nih.gov/pubmed/27676432
  • Takasato M, Er PX, Chiu HS, Maier B, Baillie GJ, Ferguson C, Parton RG, Wolvetang EJ, Roost MS, Lopes SMCDS, et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature. [Internet]. 2016;536(7615):238. accessed 2019 Sep 2. http://www.ncbi.nlm.nih.gov/pubmed/27120161
  • Morizane R, Lam AQ, Freedman BS, Kishi S, Valerius MT, Bonventre JV. Nephron organoids derived from human pluripotent stem cells model kidney development and injury.Nat Biotechnol. [Internet]. 2015 Nov;33(11):1193–200. [accessed 2019 Sep 2]. http://www.ncbi.nlm.nih.gov/pubmed/26458176
  • Camp G, Sekine K, Gerber T, Loeffler-Wirth H, Binder H, Gac M, Kanton S, Kageyama J, Damm G, Seehofer D, et al. Multilineage communication regulates human liver bud development from pluripotency. Nat Publ Gr. 2017;546(7659):533.
  • Huch M, Gehart H, van Boxtel R, Hamer K, Blokzijl F, Verstegen MMA, Ellis E, van Wenum M, Fuchs SA, de Ligt J, et al. Long-term culture of genome-stable bipotent stem cells from adult human. Cell. 2015;160(1–2):299–312.
  • McCauley HA, Wells JM. Pluripotent stem cell-derived organoids: using principles of developmental biology to grow human tissues in a dish. Development. 2017 Mar 15;144(6):958–62.
  • Harder JL, Menon R, Otto EA, Zhou J, Eddy S, Wys NL, O’Connor C, Luo J, Nair V, Cebrian C, et al. Organoid single cell profiling identifies a transcriptional signature of glomerular disease. JCI Insight. 2019 Jan 10;4(1):e122697.
  • Camp JG, Badsha F, Florio M, Kanton S, Gerber T, Wilsch-Bräuninger M, Lewitus E, Sykes A, Hevers W, Lancaster M, et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci U S A. 2015 Dec 22;112(51):15672–77.
  • Renner M, Lancaster MA, Bian S, Choi H, Ku T, Peer A, Chung K, Knoblich JA. Self-organized developmental patterning and differentiation in cerebral organoids. Embo J. [Internet]. 2017;36(10):1316–29. [accessed 2019 Aug 27]. http://www.ncbi.nlm.nih.gov/pubmed/28283582
  • Quadrato G, Nguyen T, Macosko EZ, Sherwood JL, Yang SM, Berger DR, Maria N, Scholvin J, Goldman M, Kinney JP, et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature. 2017 May 4;545(7652):48–53.
  • Eiraku M, Takata N, Ishibashi H, Kawada M, Sakakura E, Okuda S, Sekiguchi K, Adachi T, Sasai Y. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature. 2011 Apr 7;472(7341):51–58.
  • Nakano T, Ando S, Takata N, Kawada M, Muguruma K, Sekiguchi K, Saito K, Yonemura S, Eiraku M, Sasai Y. Self-formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell. [Internet]. 2012 Jun 14;10(6):771–85. [accessed 2019 Mar 14]. https://linkinghub.elsevier.com/retrieve/pii/S1934590912002421
  • Llonch S, Carido M, Ader M. Organoid technology for retinal repair. Dev Biol. [Internet]. 2018 Jan 15;433(2):132–43. [accessed 2019 Mar 14]. https://linkinghub.elsevier.com/retrieve/pii/S0012160617306620
  • Mellough CB, Collin J, Sernagor E, Wride NK, Steel DHW, Lako M. Lab generated retina: realizing the dream.Vis Neurosci. [Internet]. 2014 Sep;31(4–5):317–32. accessed 2019 Sep 4. http://www.ncbi.nlm.nih.gov/pubmed/24847731
  • Hallam D, Hilgen G, Dorgau B, Zhu L, Yu M, Bojic S, Hewitt P, Schmitt M, Uteng M, Kustermann S, et al. Human-induced pluripotent stem cells generate light responsive retinal organoids with variable and nutrient-dependent efficiency. Stem Cells. [Internet]. 2018 Oct; 36(10):1535–51. [cited 2019 Mar 14]. doi:10.1002/stem.2883.
  • Dorgau B, Felemban M, Hilgen G, Kiening M, Zerti D, Hunt NC, Doherty M, Whitfield P, Hallam D, White K. et al. Decellularised extracellular matrix-derived peptides from neural retina and retinal pigment epithelium enhance the expression of synaptic markers and light responsiveness of human pluripotent stem cell derived retinal organoids. Biomaterials. [Internet]. 2019 Apr;199:63–75. accessed 2019 Aug 13. http://www.ncbi.nlm.nih.gov/pubmed/30738336
  • Collin J, Mellough CB, Dorgau B, Przyborski S, Moreno-Gimeno I, Lako M. Using zinc finger nuclease technology to generate CRX-reporter human embryonic stem cells as a tool to identify and study the emergence of photoreceptors precursors during pluripotent stem cell differentiation. Stem Cells. 2015 Nov 27. 2016;34(2):311–21.
  • Phillips MJ, Capowski EE, Petersen A, Jansen AD, Barlow K, Edwards KL, Gamm DM. Generation of a rod-specific NRL reporter line in human pluripotent stem cells. Sci Rep. 2018;8:2370.
  • Kiselev VY, Kirschner K, Schaub MT, Andrews T, Yiu A, Chandra T, Natarajan KN, Reik W, Barahona M, Green AR, et al. SC3: consensus clustering of single-cell RNA-seq data. Nat Methods. [Internet]. 2017 May 27;14(5):483–86. [accessed 2019 Aug 13]. http://www.nature.com/articles/nmeth.4236
  • Kim S, Lowe A, Dharmat R, Lee S, Owen LA, Wang J, Shakoor A, Li Y, Morgan DJ, Hejazi AA, et al. Generation, transcriptome profiling, and functional validation of cone-rich human retinal organoids. Proc Natl Acad Sci U S A. [Internet]. 2019 May 28;116(22):10824–33. [cited 2019 Aug 13]. doi:10.1073/pnas.1901572116.
  • Daniszewski M, Senabouth A, Nguyen QH, Crombie DE, Lukowski SW, Kulkarni T, Sluch VM, Jabbari JS, Chamling X, Zack DJ, et al. Single cell RNA sequencing of stem cell-derived retinal ganglion cells. Sci Data [Internet]. 2018;5:180013. [accessed 2019 Aug 27]. http://www.ncbi.nlm.nih.gov/pubmed/29437159.
  • Langer KB, Ohlemacher SK, Phillips MJ, Fligor CM, Jiang P, Gamm DM, Meyer JS. Retinal ganglion cell diversity and subtype specification from human pluripotent stem cells. Stem Cell Rep. 2018 Apr 10;10(4):1282–93.
  • Rousso DL, Qiao M, Kagan RD, Yamagata M, Palmiter RD, Sanes JR. Two pairs of ON and OFF retinal ganglion cells are defined by intersectional patterns of transcription factor expression. Cell Rep. [Internet]. 2016;15(9):1930–44. [accessed 2019 Aug 30]. http://www.ncbi.nlm.nih.gov/pubmed/27210758
  • Puelles L. Contributions to neuroembryology of Santiago Ramon y Cajal (1852–1934) and Jorge F. Tello (1880–1958). Vol. 53. Int J Dev Biol. 2009;53:1145–60.
  • Ståhl PL, Salmén F, Vickovic S, Lundmark A, Navarro JF, Magnusson J, Giacomello S, Asp M, Westholm JO, Huss M, et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. American Association for the Advancement of Science. 2016;353:78–82.
  • Keller PJ, Ahrens MB. Visualizing whole-brain activity and development at the single-cell level using light-sheet microscopy. Neuron. Cell Press. 2015;85:462–83.