Exploring Tissue Architecture Using Spatial Transcriptomics - Biozion
Exploring Tissue Architecture Using Spatial Transcriptomics - Biozion
Exploring Tissue Architecture Using Spatial Transcriptomics - Biozion
Author manuscript
Nature. Author manuscript; available in PMC 2021 September 27.
Author Manuscript
Abstract
Author Manuscript
Deciphering the principles and mechanisms by which gene activity orchestrates complex cellular
arrangements in multicellular organisms has far-reaching implications for research in the life
sciences. Recent technological advancements in next-generation sequencing-based and imaging-
based approaches have established the potential of spatial transcriptomics to measure expression
levels of all or most genes systematically throughout tissue space, and have been adopted to
generate biological insight in neuroscience, development, plant biology, and a range of diseases
including cancer. Similar to datasets made possible by genomic sequencing and population health
surveys, the large-scale atlases generated by this technology lend themselves to exploratory
data analysis for hypothesis generation. Here, we review spatial transcriptomic technologies and
describe the repertoire of operations available for paths of analysis of the resulting data. Spatial
transcriptomics can also be deployed for hypothesis testing using experimental designs comparing
Author Manuscript
Many of the notable discoveries in the life sciences followed from the recognition
that cellular organization within tissues is intimately linked to biological function. In
developmental biology, central topics such as symmetry-breaking between daughter cells
and cell fate decisions are based on spatial relationships between cells1. In clinical settings,
histopathology is often used as a conclusive diagnostic, precisely because diseases are
characterized by abnormal spatial organization within tissues2. Infectious and inflammatory
processes can drastically change the cellular organization of tissues3. These discoveries were
supported by methods in molecular biology - including in situ hybridization4 (ISH) and
Author Manuscript
[email protected].
*Equal first authors
Author contributions: AR, DB, and IY collectively wrote the review. All authors conceptualized the figures, which GSF then created.
All authors edited and revised the manuscript.
Competing interests: The authors declare that they have no competing interests to the manuscript.
Rao et al. Page 2
The ‘omics revolution has profoundly changed our ability to characterize cells. Instead
of a few RNA or protein markers, new methods assay the full genome, transcriptome or
Author Manuscript
proteome in cells6-9. This has led to the discovery of novel cell types and cell states and
provided a more detailed understanding of biological processes in health and disease10-12.
Until recently however, these high-throughput techniques could not be applied in situ,
resulting in the loss of information about spatial relationships among the catalogued
populations of cells. To circumvent this limitation, early methods such as TomoSeq
performed transcriptomics on serial slices to reconstruct a spatial axis13-16. Similarly,
microdissection was used to manually isolate specific regions for scRNA-Seq, thus
obtaining spatially-resolved information17-23. Nanostring GeoMX digital spatial profiling
was developed to capture targeted transcripts in manually selected regions of interest24.
To reconstruct spatial relationships between neighboring cells, creative methods rely on
partial tissue dissociation25, including ProximID25-27, PICseq27 and ClumpSeq28. In another
approach, targeted mapping of a subset of genes can be used to ‘anchor’ single-cell RNA-
Author Manuscript
Seq data29-33.
While these approaches enabled the reconstruction of tissue organization, they also
highlighted the need for whole transcriptome, spatially-resolved methods. Over the past
decade, technologies have emerged that bridge the gap between traditional approaches that
retain spatial information (such as IF, ISH) and new methodologies with the ability to
concurrently query the entire transcriptome in individual cells. The inception of this new
approach of ‘spatial transcriptomics’ has facilitated novel discoveries in diverse fields from
neuroscience to development to cancer. Here, we review common spatial transcriptomic
technologies, discuss the principles of exploration of the data generated by these methods,
examine the utility of spatial transcriptomics in different experimental designs, and highlight
the promise of the technology for biological insights through integration with other
Author Manuscript
modalities.
clear-cut, and methods may incorporate elements from both categories. These diverse
technologies can be seen as converging upon a gene expression matrix (Figure 1d) capturing
the transcriptome at every spot (i.e. a pixel, a cell, or a group of cells).
with increased resolution (55µm in diameter, 100µm center-center) and sensitivity (>10k
transcripts per spot)48. Many different fields have adopted this technology, including
neuroscience49, cancer biology47,50 and developmental biology51.
Slide-Seq, another NGS-based technology, uses randomly barcoded beads deposited onto
a slide for mRNA capture52. Here, the position of each random barcode is obtained by
in situ indexing. This method has achieved high-resolution (10µm) and recently increased
sensitivity (500 transcripts per bead), which was found to be twice that of Visium for
the same surface area53. In parallel, high-definition spatial transcriptomics (HDST) also
improved the resolution by replacing the glass slide with beads deposited in wells, similar
to Slide-Seq54. More recently, the DBiT-Seq55 method has adopted microfluidics to apply
polyT barcodes to the tissue section, while Stereo-seq uses randomly barcoded DNA
nanoballs deposited in an array pattern to achieve nanoscale resolution56,57. Seq-Scope
Author Manuscript
has achieved subcellular resolution spatial barcoding and can be used to visualize nuclear
and cytoplasmic transcripts58. An innovative approach was adopted in Pixel-Seq where a
polony-derived gel oligo array was used for RNA capture resulting in up to ~200 fold
increase in resolution in comparison to existing methods59.
Common to all NGS-based methods, the spatially-barcoded RNAs are collected and
processed for sequencing. The barcode of each read is used to map the spatial position,
while the rest of the sequencing read is mapped to the genome to identify the transcript of
origin, collectively generating a gene expression matrix.
Imaging-based approaches:
Two main types of imaging-based approaches to spatial transcriptomics have been
Author Manuscript
padlock and primer design, and devised an error-robust sequencing-by-ligation method, and
was thus able to profile thousands of genes in the mouse cortex64. Other methods using
Author Manuscript
In situ hybridization (ISH) -based methods are the second category of imaging-based
methods which build on in situ hybridization technologies, whereby a target sequence is
detected by hybridization of a complementary fluorescent probe (Figure 1c). Initially limited
Author Manuscript
efforts in the community aim to improve the sensitivity and scale of these methods34,81,82.
For both ISS- and ISH-based methods, the image is processed to generate the gene
expression matrix. To obtain a cell-level matrix, the image is segmented, either manually
on small areas, or systematically using a computational approach. Watershed algorithms use
DAPI-stained nuclei as seeds and identify cell borders as regions with low RNA density83.
Although these may not correspond to true physical boundaries, but rather to the limit
between cells, they accomplish the task of assigning each mRNA to a cell. Alternatively, the
data analysis can begin at the level of individual pixels, and incorporate the gene expression
data to delineate cells84-86.
in their analysis of the dorsolateral prefrontal cortex, that led to proposed mechanisms of
genetic susceptibility to schizophrenia89. Spatial transcriptomics was also used to identify
Author Manuscript
transcriptomic dataset. Instead, there is a particular logic for how the data can be examined
and a recognition of possible outcomes with each analysis109,110.
Analyzing spatial transcriptomic data often requires the exclusion of low quality data
and initial transformations on the gene expression matrix to increase the signal-to-noise
ratio, which can be performed using analysis packages such as Giotto111, Seurat112,113,
STUtility114, and STLearn115. The total number of transcripts detected in a spot provides
a first indication of the technical and biological attributes of the data. A relatively low
number of transcripts per spot may indicate a technical artifact, such as insufficient
Author Manuscript
The normalized gene expression matrix provides the basis for initial observations at the
level of individual genes or spots (Figure 1). Revealing structure in the data, such as cell
type properties or coherent gene modules, requires further processing of the matrix. We
distinguish five classes of operations that have been used to study spatial transcriptomic
data, though more operations will undoubtedly be devised (Figure 2a). While applying any
one operation to the data may not immediately lead to insight, using the operators serially
based on the interpretation of the results at each stage can generate a ‘path’ to a result
(Figure 2b).
Cluster.
The clustering operation reveals structure in the data, most basically defining sets of
spots with similar transcriptomes or orthogonally, identifying genes with similar expression
Author Manuscript
patterns across the spots. Similarity between spots can be calculated directly between
transcriptomes using correlation or euclidean distance, or after dimensionality reduction
such as PCA, tSNE and UMAP117,118. These similarities are then used to cluster spots,
for example using k-means, Louvain or hierarchical clustering119. These clusters may
correspond to distinct regions or cell types in the tissue of study, which can then be
annotated (see ‘Characterize’). In a study on gingivitis, spots clustered according to whether
they were epithelial, connective or inflammatory120. Clustering methods were also used to
describe the tissue composition on sections of the plant A. thaliana, revealing four groups of
spots corresponding to stem, meristematic area, flower reproductive organs, and sepals and
petals90.
Gene clustering, using the same approach, can identify co-expressed gene modules
Author Manuscript
corresponding to a cell type or cell state111. In spatial transcriptomic data from the
cerebellum for example, clustering of genes identified two modules of spatially correlated
genes in Purkinje cells52. Methods to cluster genes and spots simultaneously have also
been used, including Non-negative Matrix Factorization (NMF)121,122 or factor analysis96,
where the gene expression matrix is factorized to reveal the underlying structure in spot
clusters and gene modules. In prostate tumor samples, this revealed sets of spots and
genes corresponding to cancer, stroma, and inflammation96. Currently, clustering methods
focusing on the specific features of spatial transcriptomics are being developed, such as
BayesSpace123.
Author Manuscript
Select.
Typical spatial transcriptomic datasets contain more biological information than can be
meaningfully interpreted by any single analysis. Therefore, it is usually appropriate to select
a region of interest, for example a specific layer in the brain52,53, or the interface between
tumor and microenvironment87,124. Orthogonally, one may focus the analysis on context-
specific genes, either chosen a priori from biological knowledge - most notably in imaging-
based methods which do not yet cover the whole transcriptome - or chosen from the dataset
itself - by identifying highly variable genes for example. Gene selection methods abound,
and those tailored to spatial transcriptomic data attempt to identify genes with high variance
and whose expression is not random across the tissue. Genes can be scored according
to their spatial autocorrelation (using Moran’s I or Geary’s C)125, neighbor enrichment
Author Manuscript
Score.
While the genes and spots are the primary data observations of spatial transcriptomics,
the underlying biology is such that genes are co-expressed as modules, and that spot
transcriptomes reflect a finite set of cell types and states. This is the premise of the scoring
Author Manuscript
function, which is used to summarize a cluster of similar spots as a single gene expression
profile, or - orthogonally - a coherent set of genes as a single pattern. Summarizing in this
way can identify functional properties - for example, a stress response state or infiltrating
macrophages that are spatially organized within a tumor - which might not be detectable
when analyzing spots or genes individually. Scoring can be done simply by averaging the
values of the set, or by scoring the expression relative to a null model as implemented
in the Seurat workflow113. In the brain for example, Moffitt et al. generated average cell
type expression profiles to compare spatial transcriptomics and scRNA-Seq clusters76. In
melanoma, spots were scored according to their expression of previously established gene
sets corresponding to cancer cell states45 or to Gene Ontology terms124.
Characterize.
Author Manuscript
The objects identified by operations on spatial transcriptomic data - clusters of spots and sets
of genes - must be characterized for biological understanding and interpretation. For this,
integration with other data sources and with other prior knowledge is essential. A cluster of
spots may be characterized manually when it matches a histological region, as was done in
MERFISH to annotate individual cell types in the brain76 and in pancreatic cancer samples
to annotate normal and malignant regions of the tumor46. A cluster may also be annotated
indirectly by identifying a set of marker genes and characterizing those. Specifically, a gene
set can be characterized by quantifying its overlap with an annotated gene set. This is the
basis of the Multimodal Intersection Analysis (MIA) introduced by Moncada et al.46, and of
Gene Set Enrichment Analysis (GSEA) which queries for enrichment with functional groups
Author Manuscript
ST methods with sub-cellular resolution face the inverse problem consisting of grouping
spots into organelles or cells. Seq-scope makes use of transcript annotations as spliced,
Author Manuscript
unspliced, or mitochondrial to define regions within cells58. Recent approaches have been
developed that use the local density of each RNA species to assign a cell type to each
spot84,85. Pci-Seq uses probabilistic cell typing and is able to identify cell types more
efficiently in larger tissue areas23,86. FICT, another method, integrates expression and
neighbourhood information to assign cell types141. In the case of imaging-based methods,
each DAPI-stained nucleus can be classified as a cell type according to its distance from
marker gene RNAs86.
Relate.
Given its systematic nature, spatial transcriptomics is well suited to identifying similarities,
differences and relationships between populations of genes and tissue regions. Clusters of
spots can be related by querying for expressed genes, spatial overlap, developmental or
Author Manuscript
functional relationships. For example, Stickels et al.53 identified genes that are differentially
expressed between the proximal neuropil and the soma within the hippocampus using the
different spots as replicates. Creative ways to relate the transcriptomes of clusters of spots
are borrowed from those originally developed for scRNA-Seq. RNA Velocity142 makes
use of the unspliced transcripts to infer how spots are related to each other in time, and
was applied in the cortex to map the dynamics of neuro-development53. RNA-Seq-based
Copy-number variation inference identifies chromosomal aneuploidies, which can be used to
distinguish malignant from non-malignant spots, and also identify distinct subclones143,144.
When two sets of spots are spatially adjacent, potential modes of interaction145 between
Author Manuscript
the cells can be proposed by examining their paired receptors and ligands111 using known
databases such as CellPhoneDB47,93,146 or NicheNet147.
- may lead to a novel testable hypothesis. They should also be validated independently, for
example by immunofluorescence46 or in situ hybridization76 (Figure 3a).
sections from the same sample to account for technical variability, or multiple biological
replicates per condition. The hypothesis can further be tested in model systems, in vitro or in
vivo, or in clinical data (Figure 3b).
identified and spatial smoothing is improved by averaging across spots that are not
only physically close but also similar in composition115. Another study improved the
resolution of spatial transcriptomics gene expression data by fusing it with high-resolution
histology image data148. Deep learning has also been used to predict cell type annotations
from gene expression and histology, outperforming annotations predicted from either
modality alone149. With the increase in transcriptomic data available for training, machine
learning algorithms have also been used to predict gene expression from histopathology
transcriptomics with such machine learning approaches may improve the interpretability
of histopathology and its use in clinical decision making to guide treatment and inform
prognosis.
At subcellular resolution, the spatial organization of chromatin may provide clues into the
regulation of gene expression in various contexts. DNA seqFISH integrated with RNA
seqFISH and multiplexed immunofluorescence revealed that active gene loci are located on
the surface of nuclear bodies and zone interfaces in embryonic stem cells152. Integrating
spatial transcriptomic datasets with high-throughput imaging of genome in situ and the
spatial distribution of histone marks within a tissue will be extremely valuable153-155.
Recently, spatial mapping of genome organization with concurrent DNA sequencing within
intact tissue has been made feasible156. This suggests that the goal of combining spatial
Author Manuscript
genome sequencing with in situ transcriptomic profiling may be within reach, deepening our
understanding how genome organization and function are encoded155.
Augmenting gene expression data with a complementary modality like protein-co detection
can also shed light into processes that spatial transcriptomics does not capture, such as post-
translational modification and sub-cellular localization of proteins and their dysregulation
in disease. Targeted protein co-detection performed alongside spatial transcriptomics can
be achieved using immunostaining on the same tissue section, as enabled by Visium48. A
novel imaging cytometry based approach was used to simultaneously detect transcripts and
proteins in breast cancer tissue samples157. DBiT-Seq allows for the co-mapping of mRNA
and proteins in the tissue using antibody-derived DNA tags, as is done in CITE-Seq158.
High-throughput spatial methods for protein detection such as MIBI, CODEX, t-cyCIF as
Author Manuscript
well as mass spectrometry and barcode based approaches provide an unparalleled snapshot
of the proteome within the tissue section159-164. Technological advances that allow the
integration of these high-throughput proteomics methods with spatial transcriptomics will
tremendously improve our ability to study tissue complexity.
Outlook
The spatial transcriptomics field is growing at an exponential pace, with daily releases of
technologies and datasets. The challenges faced by current spatial transcriptomic methods
- including the limits to resolution and sensitivity, as well as throughput and accessibility -
are being rapidly overcome. Spatial transcriptomics methods are being made compatible
with paraffin-embedded tissues, opening the door to retrospective analyses of samples
collected over decades in biobanks48,70,165,166. With future innovations, it may be possible
Author Manuscript
to systematically assay larger tissue areas for the reconstruction of 3D organ- or organism-
level atlases, and to visualize transcriptome-wide gene expression changes as they unfold
over time. In addition to overcoming these technological challenges, future work will require
the development of new computational tools and creative analytical thinking. Together,
these will enable data exploration to identify ‘spatial patterns’ - a central feature of spatial
transcriptomic datasets - and reveal insights into the underlying biology.
As we speculate about the future milestones of the field, the human genome project
may serve as a useful parallel. The initial draft of the human genome was published in
Author Manuscript
2001167,168 and provided a reference to study the sources and consequences of genetic
variation. However, the function and regulation of the different regions of the genome
are still under active investigation. In spatial transcriptomics, future projects may similarly
benefit from a reference from which to study distinct conditions. However, mapping the
expression level of every gene in space will only be the first step to elucidating organizing
principles of tissue biology. It is the coupling of these high-resolution cellular atlases with
hypothesis-free inquiries that will enable new insight, and reveal the salient features of tissue
architecture in physiology and disease.
A key challenge for the field will be to iteratively build a model of how multicellular
spatial patterns emerge from cell-level properties. Independent of spatial transcriptomic
technologies, implementing a simple principle - that each cell is overall most similar to its
Author Manuscript
neighbors - was sufficient to recover complex spatial patterns in the Drosophila embryo169.
Building on this idea, the exploration of spatial transcriptomic datasets will enable us
to uncover the fundamental principles that guide our modeling of tissue-level spatial
organization and will facilitate the study of the mechanistic basis of these patterns and their
consequences. These deeper biological insights will extend the level of understanding from
simple tissues to more complex structures, including developing organisms and diseased
tissues, bringing us closer to conquering the spatial frontier.
Acknowledgements:
We thank Felicia Kuperwaser, Andrew Pountain, Bo Xia, and other members of the Yanai lab, as well as Mark
Phillips for critical reading and feedback. We thank the students of the exploratory data analysis course at
NYU Langone Medical Center. IY was supported by grants from the NIH (R01AI143290) and the Lowenstein
Author Manuscript
Foundation, and DB was supported by the NIH (F30CA257400). We also thank the three anonymous reviewers for
important suggestions.
References:
1. Barresi MJF & Gilbert SF Developmental Biology. (Sinauer Associates, Incorporated, 2019).
2. Damjanov I & McCue PA Histopathology: A Color Atlas and Textbook. (Lippincott Williams &
Wilkins, 1996).
3. Safai B & Good RA Immunodermatology. (Springer Science & Business Media, 2013).
4. Lehmann R & Tautz D In Situ Hybridization to RNA. in Methods in Cell Biology vol. 44 575–598
(Academic Press, 1994). [PubMed: 7535885]
5. Swanson PEFoundations of Immunohistochemistry. Am. J. Clin. Pathol90, 333–339 (1988).
[PubMed: 3046324]
6. Mincarelli L, Lister A, Lipscombe J & Macaulay IC Defining Cell Identity with Single-Cell Omics.
Proteomics 18, e1700312 (2018). [PubMed: 29644800]
Author Manuscript
7. Macaulay IC, Ponting CP & Voet T Single-Cell Multiomics: Multiple Measurements from Single
Cells. Trends Genet. 33, 155–168 (2017). [PubMed: 28089370]
8. Liu XZ, Peng YZ & Shen ZA [Research advances on application of single-cell RNA sequencing in
islet cell biology]. Zhonghua Shao Shang Za Zhi 36, 1208–1212 (2020). [PubMed: 33379859]
9. Xia B & Yanai I A periodic table of cell types. Development 146, (2019).
10. Zheng G, Xie Z-Y, Wang P, Wu Y-F & Shen H-Y Recent advances of single-cell RNA
sequencing technology in mesenchymal stem cell research. World J. Stem Cells 12, 438–447
(2020). [PubMed: 32742561]
11. Lawson DA, Kessenbrock K, Davis RT, Pervolarakis N & Werb Z Tumour heterogeneity and
metastasis at single-cell resolution. Nat. Cell Biol 20, 1349–1360 (2018). [PubMed: 30482943]
Author Manuscript
12. Papalexi E & Satija R Single-cell RNA sequencing to explore immune cell heterogeneity. Nat. Rev.
Immunol 18, 35–45 (2018). [PubMed: 28787399]
13. Combs PA & Eisen MB Sequencing mRNA from cryo-sliced Drosophila embryos to determine
genome-wide spatial patterns of gene expression. PLoS One 8, e71820 (2013). [PubMed:
23951250]
14. Junker JPet al.Genome-wide RNA Tomography in the zebrafish embryo. Cell159, 662–675 (2014).
[PubMed: 25417113]
15. Lacraz GPAet al.Tomo-Seq Identifies SOX9 as a Key Regulator of Cardiac Fibrosis During
Ischemic Injury. Circulation136, 1396–1409 (2017). [PubMed: 28724751]
16. van den Brink SCet al.Single-cell and spatial transcriptomics reveal somitogenesis in gastruloids.
Nature582, 405–409 (2020). [PubMed: 32076263]
17. Nichterwitz Set al.Laser capture microscopy coupled with Smart-seq2 for precise spatial
transcriptomic profiling. Nat. Commun7, 12139 (2016). [PubMed: 27387371]
18. Nichterwitz S, Benitez JA, Hoogstraaten R, Deng Q & Hedlund E LCM-Seq: A Method for Spatial
Author Manuscript
Transcriptomic Profiling Using Laser Capture Microdissection Coupled with PolyA-Based RNA
Sequencing. Methods in Molecular Biology 95–110 (2018) doi:10.1007/978-1-4939-7213-5_6.
19. Aguila Jet al.Spatial transcriptomics and in silico random pooling identify novel markers of
vulnerable and resistant midbrain dopamine neurons. doi:10.1101/334417.
20. Moor AEet al.Spatial Reconstruction of Single Enterocytes Uncovers Broad Zonation along the
Intestinal Villus Axis. Cell175, 1156–1167.e15 (2018). [PubMed: 30270040]
21. Chen Jet al.Spatial transcriptomic analysis of cryosectioned tissue samples with Geo-seq. Nat.
Protoc12, 566–580 (2017). [PubMed: 28207000]
22. Tirosh Iet al.Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-
seq. Science352, 189–196 (2016). [PubMed: 27124452]
23. Peng Get al.Molecular architecture of lineage allocation and tissue organization in early mouse
embryo. Nature572, 528–532 (2019). [PubMed: 31391582]
24. Geiss GKet al.Direct multiplexed measurement of gene expression with color-coded probe pairs.
Nat. Biotechnol26, 317–325 (2008). [PubMed: 18278033]
Author Manuscript
25. Halpern KBet al.Paired-cell sequencing enables spatial gene expression mapping of liver
endothelial cells. Nat. Biotechnol36, 962–970 (2018). [PubMed: 30222169]
26. Boisset J-Cet al.Mapping the physical network of cellular interactions. Nat. Methods15, 547–553
(2018). [PubMed: 29786092]
27. Giladi Aet al.Dissecting cellular crosstalk by sequencing physically interacting cells. Nat.
Biotechnol38, 629–637 (2020). [PubMed: 32152598]
28. Manco Ret al.Clump sequencing exposes the spatial expression programs of intestinal secretory
cells. Cold Spring Harbor Laboratory 2020.08.05.237917 (2020) doi:10.1101/2020.08.05.237917.
29. Satija R, Farrell JA, Gennert D, Schier AF & Regev A Spatial reconstruction of single-cell gene
expression data. Nature Biotechnology vol. 33 495–502 (2015).
30. Achim Ket al.High-throughput spatial mapping of single-cell RNA-seq data to tissue of origin. Nat.
Biotechnol33, 503–509 (2015). [PubMed: 25867922]
31. Pettit J-Bet al.Identifying cell types from spatially referenced single-cell expression datasets. PLoS
Comput. Biol10, e1003824 (2014). [PubMed: 25254363]
Author Manuscript
32. Karaiskos Net al.The embryo at single-cell transcriptome resolution. Science358, 194–199 (2017).
[PubMed: 28860209]
33. Cang Z & Nie Q Inferring spatial and signaling relationships between cells from single cell
transcriptomic data. Nat. Commun 11, 2084 (2020). [PubMed: 32350282]
34. Zhuang XSpatially resolved single-cell genomics and transcriptomics by imaging. Nat. Methods18,
18–22 (2021). [PubMed: 33408406]
35. Larsson L, Frisén J & Lundeberg J Spatially resolved transcriptomics adds a new dimension to
genomics. Nat. Methods 18, 15–18 (2021). [PubMed: 33408402]
36. Crosetto N, Bienko M & van Oudenaarden A Spatially resolved transcriptomics and beyond. Nat.
Rev. Genet 16, 57–66 (2015). [PubMed: 25446315]
Author Manuscript
37. Moor AE & Itzkovitz S Spatial transcriptomics: paving the way for tissue-level systems biology.
Curr. Opin. Biotechnol 46, 126–133 (2017). [PubMed: 28346891]
38. Asp M, Bergenstråhle J & Lundeberg J Spatially Resolved Transcriptomes-Next Generation Tools
for Tissue Exploration. Bioessays 42, e1900221 (2020). [PubMed: 32363691]
39. Waylen LN, Nim HT, Martelotto LG & Ramialison M From whole-mount to single-cell spatial
assessment of gene expression in 3D. Commun Biol 3, 602 (2020). [PubMed: 33097816]
40. Teves JM & Won KJ Mapping Cellular Coordinates through Advances in Spatial Transcriptomics
Technology. Mol. Cells 43, 591–599 (2020). [PubMed: 32507771]
41. Ståhl PLet al.Visualization and analysis of gene expression in tissue sections by spatial
transcriptomics. Science353, 78–82 (2016). [PubMed: 27365449] This paper was the first to
perform array-based spatial transcriptomics, using positional barcodes at a resolution of 200µm,
and demonstrated the approach on the mouse olfactory bulb.
42. Jemt Aet al.An automated approach to prepare tissue-derived spatially barcoded RNA-sequencing
libraries. Sci. Rep6, 37137 (2016). [PubMed: 27849009]
Author Manuscript
43. Salmén Fet al.Barcoded solid-phase RNA capture for Spatial Transcriptomics profiling in
mammalian tissue sections. Nat. Protoc13, 2501–2534 (2018). [PubMed: 30353172]
44. Thrane K, Eriksson H, Maaskola J, Hansson J & Lundeberg J Spatially Resolved Transcriptomics
Enables Dissection of Genetic Heterogeneity in Stage III Cutaneous Malignant Melanoma. Cancer
Res. 78, 5970–5979 (2018). [PubMed: 30154148]
45. Baron Met al.The Stress-Like Cancer Cell State Is a Consistent Component of Tumorigenesis. Cell
Syst (2020) doi:10.1016/j.cels.2020.08.018.
46. Moncada Ret al.Integrating microarray-based spatial transcriptomics and single-cell RNA-seq
reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat. Biotechnol38, 333–342
(2020). [PubMed: 31932730] This paper performed array-based ST and introduced multimodal
intersection analysis to study pancreatic cancer and identify cell type co-localizations.
47. Ji ALet al.Multimodal Analysis of Composition and Spatial Architecture in Human Squamous Cell
Carcinoma. Cell182, 497–514.e22 (2020). [PubMed: 32579974] This paper used array-based ST
to identify genes at the leading edge of the tumor in squamous cell carcinoma.
Author Manuscript
58. Cho C-Set al.Seq-Scope: Submicrometer-resolution spatial transcriptomics for single cell and
subcellular studies. bioRxiv (2021) doi:10.1101/2021.01.25.427807.
Author Manuscript
59. Fu Xet al.Continuous Polony Gels for Tissue Mapping with High Resolution and
RNA Capture Efficiency. Cold Spring Harbor Laboratory 2021.03.17.435795 (2021)
doi:10.1101/2021.03.17.435795.
60. Ke Ret al.In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods10,
857–860 (2013). [PubMed: 23852452] This paper first performed in situ sequencing, mapping the
expression of 31 transcripts using 4-base reads in breast cancer.
61. Darmanis Set al.Single-Cell RNA-Seq Analysis of Infiltrating Neoplastic Cells at the Migrating
Front of Human Glioblastoma. Cell Rep. 21, 1399–1410 (2017). [PubMed: 29091775]
62. Carow Bet al.Spatial and temporal localization of immune transcripts defines hallmarks and
diversity in the tuberculosis granuloma. Nat. Commun10, 1823 (2019). [PubMed: 31015452]
63. Tiklová Ket al.Single-cell RNA sequencing reveals midbrain dopamine neuron diversity emerging
during mouse brain development. Nat. Commun10, 581 (2019). [PubMed: 30718509]
64. Wang Xet al.Three-dimensional intact-tissue sequencing of single-cell transcriptional states.
Science361, (2018).
Author Manuscript
65. Chen X, Sun Y-C, Church GM, Lee JH & Zador AM Efficient in situ barcode sequencing using
padlock probe-based BaristaSeq. Nucleic Acids Res. 46, e22 (2018). [PubMed: 29190363]
66. Chen Xet al.High-Throughput Mapping of Long-Range Neuronal Projection Using In Situ
Sequencing. Cell179, 772–786.e19 (2019). [PubMed: 31626774]
67. Gyllborg Det al.Hybridization-based in situ sequencing (HybISS) for spatially resolved
transcriptomics in human and mouse brain tissue. Nucleic Acids Res. 48, e112 (2020). [PubMed:
32990747]
68. Fürth D, Hatini V & Lee JH In Situ Transcriptome Accessibility Sequencing (INSTA-seq). Cold
Spring Harbor Laboratory 722819 (2019) doi:10.1101/722819.
69. Alon Set al.Expansion sequencing: Spatially precise in situ transcriptomics in intact biological
systems. Science doi:10.1126/science.aax2656.
70. Lee JHet al.Highly multiplexed subcellular RNA sequencing in situ. Science343, 1360–1363
(2014). [PubMed: 24578530] This paper introduced an untargeted in situ sequencing method,
FISSEQ, that generated 30 base reads from 8102 genes demonstrated in human primary
Author Manuscript
fibroblasts.
71. Codeluppi Set al.Spatial organization of the somatosensory cortex revealed by osmFISH. Nat.
Methods15, 932–935 (2018). [PubMed: 30377364]
72. Chen KH, Boettiger AN, Moffitt JR, Wang S & Zhuang X RNA imaging. Spatially resolved,
highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015). [PubMed:
25858977]
73. Moffitt JRet al.High-throughput single-cell gene-expression profiling with multiplexed error-robust
fluorescence in situ hybridization. Proc. Natl. Acad. Sci. U. S. A113, 11046–11051 (2016).
[PubMed: 27625426]
74. Wang G, Moffitt JR & Zhuang X Multiplexed imaging of high-density libraries of RNAs with
MERFISH and expansion microscopy. Sci. Rep 8, 4847 (2018). [PubMed: 29555914]
75. Xia C, Babcock HP, Moffitt JR & Zhuang X Multiplexed detection of RNA using MERFISH and
branched DNA amplification. Sci. Rep 9, 7721 (2019). [PubMed: 31118500]
76. Moffitt JRet al.Molecular, spatial, and functional single-cell profiling of the hypothalamic preoptic
region. Science362, (2018). This paper combines MERFISH, an in situ hybridization-based
Author Manuscript
81. Crosetto N, Bienko M & van Oudenaarden A Spatially resolved transcriptomics and beyond. Nat.
Rev. Genet 16, 57–66 (2015). [PubMed: 25446315]
82. Asp M, Bergenstråhle J & Lundeberg J Spatially Resolved Transcriptomes-Next Generation Tools
for Tissue Exploration. Bioessays 42, e1900221 (2020). [PubMed: 32363691]
83. Najman L & Schmitt M Watershed of a continuous function. Signal Processing (1994)
doi:10.1016/0165-1684(94)90059-0.
84. Park JSegmentation-free Inference of Cell Types from in Situ Transcriptomics Data. (2020).
85. Littman Ret al.JSTA: joint cell segmentation and cell type annotation for spatial transcriptomics.
doi:10.1101/2020.09.18.304147.
86. Qian Xet al.Probabilistic cell typing enables fine mapping of closely related cell types in situ. Nat.
Methods17, 101–106 (2020). [PubMed: 31740815]
87. BRAIN Initiative Cell Census Network (BICCN)et al.A multimodal cell census and atlas of the
mammalian primary motor cortex. Cold Spring Harbor Laboratory 2020.10.19.343129 (2020)
doi:10.1101/2020.10.19.343129.
Author Manuscript
88. Kebschull JMet al.Cerebellar nuclei evolved by repeatedly duplicating a conserved cell-type set.
Science370, (2020).
89. Maynard KRet al.Transcriptome-scale spatial gene expression in the human dorsolateral prefrontal
cortex. doi:10.1101/2020.02.28.969931.
90. Giacomello Set al.Spatially resolved transcriptome profiling in model plant species. Nat Plants3,
17061 (2017). [PubMed: 28481330]
91. Chen Het al.Dissecting Mammalian Spermatogenesis Using Spatial Transcriptomics.
doi:10.1101/2020.10.17.343335.
92. Fawkner-Corbett Det al.Spatiotemporal analysis of human intestinal development at single-cell
resolution. Cell184, 810–826.e23 (2021). [PubMed: 33406409] Spatial transcriptomics was
performed on the intestine at various stages of development, revealing how cell types are organized
along the crypt-villus axis.
93. Garcia-Alonso Let al.Mapping the temporal and spatial dynamics of the human endometrium in
vivo and in vitro. doi:10.1101/2021.01.02.425073.
Author Manuscript
94. Rozenblatt-Rosen Oet al.The Human Tumor Atlas Network: Charting Tumor Transitions across
Space and Time at Single-Cell Resolution. Cell181, 236–249 (2020). [PubMed: 32302568]
95. Lein ESet al.Genome-wide atlas of gene expression in the adult mouse brain. Nature445, 168–176
(2007). [PubMed: 17151600]
96. Berglund Eet al.Spatial maps of prostate cancer transcriptomes reveal an unexplored landscape of
heterogeneity. Nat. Commun9, 2419 (2018). [PubMed: 29925878]
97. Wang Y, Ma S & Ruzzo WL Spatial modeling of prostate cancer metabolic gene expression
reveals extensive heterogeneity and selective vulnerabilities. Sci. Rep 10, 3490 (2020). [PubMed:
32103057]
98. Hwang WLet al.Single-nucleus and spatial transcriptomics of archival pancreatic cancer reveals
multi-compartment reprogramming after neoadjuvant treatment. doi:10.1101/2020.08.25.267336.
99. Smith EA & Hodges HC The Spatial and Genomic Hierarchy of Tumor Ecosystems Revealed by
Single-Cell Technologies. Trends Cancer Res. 5, 411–425 (2019).
100. Navarro JFet al.Spatial Transcriptomics Reveals Genes Associated with Dysregulated
Author Manuscript
Mitochondrial Functions and Stress Signaling in Alzheimer Disease. iScience23, 101556 (2020).
[PubMed: 33083725]
101. Chen W-Tet al.Spatial Transcriptomics and In Situ Sequencing to Study Alzheimer’s Disease.
Cell182, 976–991.e19 (2020). [PubMed: 32702314]
102. Maniatis Set al.Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis.
Science364, 89–93 (2019). [PubMed: 30948552]
103. Ma Fet al.Single Cell and Spatial Transcriptomics Defines the Cellular
Architecture of the Antimicrobial Response Network in Human Leprosy Granulomas.
doi:10.1101/2020.12.01.406819.
104. Boyd DFet al.Exuberant fibroblast activity compromises lung function via ADAMTS4.
Nature587, 466–471 (2020). [PubMed: 33116313]
Author Manuscript
105. Janosevic Det al.The orchestrated cellular and molecular responses of the kidney to endotoxin
define a precise sepsis timeline. Elife10, (2021).
106. Carlberg Ket al.Exploring inflammatory signatures in arthritic joint biopsies with Spatial
Transcriptomics. Scientific Reports vol. 9 (2019).
107. Vickovic Set al.Three-dimensional spatial transcriptomics uncovers cell type dynamics in the
rheumatoid arthritis synovium. doi:10.1101/2020.12.10.420463.
108. Tukey JWExploratory Data Analysis. (1970). John Tukey established the field of exploratory data
analysis as an approach to discover trends prior to testing for any particular model.
109. Yanai I & Lercher M What is the question? Genome Biol. 20, 1–5 (2019). [PubMed: 30606230]
110. Yanai I & Lercher M The data-hypothesis conversation. Genome Biol. 22, 1–6 (2021). [PubMed:
33397451]
111. Dries Ret al.Giotto: a toolbox for integrative analysis and visualization of spatial expression
data. Genome Biol. 22, 1–31 (2021). [PubMed: 33397451] Giotto enables analysis of spatial
transcriptomics data, including normalization, identification of spatially variable genes and cell
Author Manuscript
117. Zhou B & Jin W Visualization of Single Cell RNA-Seq Data Using t-SNE in R. Methods Mol.
Biol 2117, 159–167 (2020). [PubMed: 31960377]
118. Becht Eet al.Dimensionality reduction for visualizing single-cell data using UMAP. Nat.
Biotechnol (2018) doi:10.1038/nbt.4314.
119. Kiselev VY, Andrews TS & Hemberg M Challenges in unsupervised clustering of single-cell
RNA-seq data. Nat. Rev. Genet 20, 273–282 (2019). [PubMed: 30617341]
120. Lundmark Aet al.Gene expression profiling of periodontitis-affected gingival tissue by spatial
transcriptomics. Sci. Rep8, 9370 (2018). [PubMed: 29921943]
121. Lee DD & Seung HS Learning the parts of objects by non-negative matrix factorization. Nature
401, 788–791 (1999). [PubMed: 10548103]
122. Gao Y & Church G Improving molecular cancer class discovery through sparse non-negative
matrix factorization. Bioinformatics 21, 3970–3975 (2005). [PubMed: 16244221]
123. Zhao Eet al.BayesSpace enables the robust characterization of spatial gene expression
architecture in tissue sections at increased resolution. doi:10.1101/2020.09.04.283812.
Author Manuscript
124. Hunter MV, Moncada R, Weiss JM, Yanai I & White RM Spatial transcriptomics reveals the
architecture of the tumor/microenvironment interface. doi:10.1101/2020.11.05.368753.
125. Moran PAPNotes on continuous stochastic phenomena. Biometrika37, 17–23 (1950). [PubMed:
15420245]
126. Vandenbon A & Diez D A clustering-independent method for finding differentially expressed
genes in single-cell transcriptome data. Nat. Commun 11, 4318 (2020). [PubMed: 32859930]
127. Edsgärd D, Johnsson P & Sandberg R Identification of spatial expression trends in single-cell
gene expression data. Nat. Methods 15, 339–342 (2018). [PubMed: 29553578] Trendsceek is a
computational method for identifying spatially variable genes using marked point processes, and
was tested on mouse olfactory bulb and hippocampus regions and human breast cancer tissue.
Author Manuscript
128. Illian J, Penttinen A, Stoyan H & Stoyan D Statistical Analysis and Modelling of Spatial Point
Patterns. (John Wiley & Sons, 2008).
129. Svensson V, Teichmann SA & Stegle O SpatialDE: identification of spatially variable genes.
Nat. Methods 15, 343–346 (2018). [PubMed: 29553579] SpatialDE describes a statistical test for
identifying genes with a spatial pattern using a non-parametric regression based approach.
130. Sun S, Zhu J & Zhou X Statistical analysis of spatial expression patterns for spatially resolved
transcriptomic studies. Nat. Methods 17, 193–200 (2020). [PubMed: 31988518] This paper
reports a new computational method that uses a generalized linear spatial model to identify
spatially variable genes from spatial transcriptomics datasets.
131. Ashburner Met al.Gene ontology: tool for the unification of biology. The Gene Ontology
Consortium. Nat. Genet25, 25–29 (2000). [PubMed: 10802651]
132. Subramanian Aet al.Gene set enrichment analysis: A knowledge-based approach for interpreting
genome-wide expression profiles. Proc. Natl. Acad. Sci. U. S. A102, 15545–15550 (2005).
[PubMed: 16199517]
Author Manuscript
133. Kanehisa M & Goto S KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28,
27–30 (2000). [PubMed: 10592173]
134. Elosua M, Nieto P, Mereu E, Gut I & Heyn H SPOTlight: Seeded NMF regression to Deconvolute
Spatial Transcriptomics Spots with Single-Cell Transcriptomes. doi:10.1101/2020.06.03.131334.
135. Andersson Aet al.Single-cell and spatial transcriptomics enables probabilistic inference of cell
type topography. Commun Biol3, 565 (2020). [PubMed: 33037292]
136. Kleshchevnikov Vet al.Comprehensive mapping of tissue cell architecture via integrated single
cell and spatial transcriptomics. doi:10.1101/2020.11.15.378125.
137. Cable DMet al.Robust decomposition of cell type mixtures in spatial transcriptomics.
doi:10.1101/2020.05.07.082750.
138. Su J & Song Q DSTG: Deconvoluting Spatial Transcriptomics Data through Graph-based
Artificial Intelligence. doi:10.1101/2020.10.20.347195.
139. Biancalani Tet al.Deep learning and alignment of spatially-resolved whole transcriptomes of
single cells in the mouse brain with Tangram. bioRxiv (2020) doi:10.1101/2020.08.29.272831.
Author Manuscript
140. Dong R & Yuan G-C SpatialDWLS: accurate deconvolution of spatial transcriptomic data.
doi:10.1101/2021.02.02.429429.
141. Teng H, Yuan Y & Bar-Joseph Z Cell Type Assignments for Spatial Transcriptomics Data.
bioRxiv 2021.02.25.432887 (2021) doi:10.1101/2021.02.25.432887.
142. La Manno Get al.RNA velocity of single cells. Nature560, 494–498 (2018). [PubMed: 30089906]
143. Patel APet al.Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma.
Science344, 1396–1401 (2014). [PubMed: 24925914]
144. Elyanow R, Zeira R, Land M & Raphael B STARCH: Copy number and clone inference from
spatial transcriptomics data. Phys. Biol (2020) doi:10.1088/1478-3975/abbe99.
145. Armingol E, Officer A, Harismendy O & Lewis NE Deciphering cell-cell interactions and
communication from gene expression. Nat. Rev. Genet 22, 71–88 (2021). [PubMed: 33168968]
146. Efremova M, Vento-Tormo M, Teichmann SA & Vento-Tormo R CellPhoneDB: inferring cell-cell
communication from combined expression of multi-subunit ligand-receptor complexes. Nat.
Protoc 15, 1484–1506 (2020). [PubMed: 32103204]
Author Manuscript
147. Browaeys R, Saelens W & Saeys Y NicheNet: modeling intercellular communication by linking
ligands to target genes. Nat. Methods 17, 159–162 (2020). [PubMed: 31819264]
148. Bergenstråhle Let al.Super-resolved spatial transcriptomics by deep data fusion. Cold Spring
Harbor Laboratory 2020.02.28.963413 (2020) doi:10.1101/2020.02.28.963413.
149. Tan X, Su A, Tran M & Nguyen Q SpaCell: integrating tissue morphology and spatial gene
expression to predict disease cells. doi:10.1101/837211.
150. He Bet al.Integrating spatial gene expression and breast tumour morphology via deep learning.
Nat Biomed Eng4, 827–834 (2020). [PubMed: 32572199] This study develops a deep learning
method to predict gene expression from histopathology images of breast cancer patients.
151. Levy-Jurgenson A, Tekpli X, Kristensen VN & Yakhini Z Spatial transcriptomics inferred from
pathology whole-slide images links tumor heterogeneity to survival in breast and lung cancer.
Author Manuscript
[PubMed: 29698648]
158. Stoeckius Met al.Simultaneous epitope and transcriptome measurement in single cells. Nat.
Methods14, 865–868 (2017). [PubMed: 28759029]
159. Angelo Met al.Multiplexed ion beam imaging of human breast tumors. Nat. Med20, 436–442
(2014). [PubMed: 24584119]
160. Keren Let al.A Structured Tumor-Immune Microenvironment in Triple Negative Breast Cancer
Revealed by Multiplexed Ion Beam Imaging. Cell174, 1373–1387.e19 (2018). [PubMed:
30193111]
161. Piehowski PDet al.Automated mass spectrometry imaging of over 2000 proteins from tissue
sections at 100-µm spatial resolution. Nat. Commun11, 8 (2020). [PubMed: 31911630]
162. Lin J-Ret al.Highly multiplexed immunofluorescence imaging of human tissues and tumors using
t-CyCIF and conventional optical microscopes. Elife7, (2018).
163. Goltsev Yet al.Deep Profiling of Mouse Splenic Architecture with CODEX Multiplexed Imaging.
Cell174, 968–981.e15 (2018). [PubMed: 30078711]
Author Manuscript
164. Kohman RE & Church GM Fluorescent in situ sequencing of DNA barcoded antibodies. Cold
Spring Harbor Laboratory 2020.04.27.060624 (2020) doi:10.1101/2020.04.27.060624.
165. Liu Y, Enninful A, Deng Y & Fan R Spatial transcriptome sequencing of FFPE
tissues at cellular level. Cold Spring Harbor Laboratory 2020.10.13.338475 (2020)
doi:10.1101/2020.10.13.338475.
166. Nagarajan MB, Tentori AM, Zhang WC, Slack FJ & Doyle PS Spatially resolved and multiplexed
MicroRNA quantification from tissue using nanoliter well arrays. Microsyst Nanoeng 6, 51
(2020). [PubMed: 32419951]
167. Lander ESet al.Initial sequencing and analysis of the human genome. Nature409, 860–921 (2001).
[PubMed: 11237011]
168. Venter JCet al.The sequence of the human genome. Science291, 1304–1351 (2001). [PubMed:
11181995]
169. Nitzan M, Karaiskos N, Friedman N & Rajewsky N Gene expression cartography. Nature 576,
132–137 (2019). [PubMed: 31748748] The NovaSparc method reconstructs spatial organization
from single-cell RNA-Seq data by positing that cells are overall most similar to their neighbors
Author Manuscript
in systems that are inherently symmetrical, and was demonstrated in liver lobules, intestinal villi,
and whole Drosophila embryos.
170. Xia C, Fan J, Emanuel G, Hao J & Zhuang X Spatial transcriptome profiling by MERFISH
reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression. Proc.
Natl. Acad. Sci. U. S. A 116, 19490–19499 (2019). [PubMed: 31501331]
171. Joglekar Aet al.A spatially resolved brain region- and cell type-specific isoform atlas of the
postnatal mouse brain. Nat. Commun12, 463 (2021). [PubMed: 33469025]
172. Frieda KLet al.Synthetic recording and in situ readout of lineage information in single cells.
Nature541, 107–111 (2017). [PubMed: 27869821]
173. Rodriques SGet al.Slide-seq: A scalable technology for measuring genome-wide expression at
high spatial resolution. Science363, 1463–1467 (2019). [PubMed: 30923225]
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
Box 1:
Author Manuscript
Sequence information:
In NGS-based and ISS-based methods, the cDNA sequence itself is a read out, enabling
the detection of splice isoforms58,56,171,56 as well as single nucleotide variants and point
mutations60. When integrated with the gene expression matrix, this data can assist with
reconstructing a time-course - using RNA Velocity53 or lineage tracing172.
Sensitivity:
ISH-based methods are highly sensitive, recently reaching 80% detection efficiency
relative to the gold standard smFISH170. Sensitivity of the NGS-based methods is
significantly lower and remains inferior to scRNA-Seq, but is rapidly improving to
~100 unique transcripts per square µm54,58,59,173. There is generally a tradeoff between
Author Manuscript
sensitivity and gene throughput, as seen in the higher sensitivity of targeted ISS-based
methods64 relative to the unbiased methods70.
Resolution:
The resolution of in situ methods is limited only by the optical diffraction limit, and
with expansion microscopy has reached ~100nm80,170. These methods are therefore
well-suited to questions concerning sub-cellular organization. NGS-based methods are
limited by the diameter of spots, but their resolution has rapidly increased since the
original method41, recently reaching ~1µm58,59.
Area size:
The in situ methods can span a wide range of sizes, although there is a tradeoff
between tissue size and imaging time73. The NGS-based methods on the other hand
Author Manuscript
are standardized, with arrays on the order of ten square mm (6 for Visium), which may be
inappropriate for smaller or larger samples.
Feasibility:
While these technologies are extremely powerful, there are obstacles to their widespread
adoption, including access to single molecule imaging for in situ methods, as well as
manufacturing for the capture arrays. Commercialization has facilitated access in some
cases.
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript
location in a lattice of spots. b, In situ sequencing approaches directly read out the
transcript sequence within the tissue. c, In situ hybridization methods detect target
sequences by hybridization of complementary fluorescent probes. d, The product of spatial
transcriptomics is the gene expression matrix - where the rows and columns correspond to
genes and locations.
Author Manuscript
Author Manuscript
for analysis. Composition: Spots are scored for cell type-specific gene expression profiles
from scRNA-Seq data and characterized to identify the composition of the tissue region.
Co-localization: Co-varying genes are identified by clustering and spots are scored for
the expression of these gene sets to identify a pattern of overlapping spatial expression. A
co-localization is described by relating the distance between these spots. Communication:
Transcriptionally similar spots are identified by clustering and characterized according to
their resident cell types. A subset of receptor and ligand pairs are selected for analysis.
Receptors and ligands expressed in cell type α and cell type β, respectively, suggests a
relationship between them.
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript