You are on page 1of 14

Review Article

Spatially resolved transcriptomics:


advances and applications
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

Honglin Duana, Tao Chenga,b,c,*, Hui Chenga,b,c,*


a
State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center
for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking
Union Medical College, Tianjin, China; bCenter for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China;
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

c
Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin, China

Abstract
Spatial transcriptomics, which is capable of both measuring all gene activity in a tissue sample and mapping where this activity
occurs, is vastly improving our understanding of biological processes and disease. The field has expanded rapidly in recent
years, and the development of several new technologies has resulted in spatially resolved transcriptomics (SRT) becoming highly
multiplexed, high-resolution, and high-throughput. Here, we summarize and compare the major methods of SRT, including imaging-
based methods, sequencing-based methods, and in situ sequencing methods. We also highlight some typical applications of
SRT in neuroscience, cancer biology, developmental biology, and hematology. Finally, we discuss future possibilities for improving
spatially resolved transcriptomic methods and the expected applications of such methods, especially in the adult bone marrow,
anticipating that new developments will unlock the full potential of spatially resolved multi-omics in both biological research and
the clinic.
Key Words: In situ hybridization; In situ sequencing; Spatially resolved multi-omics; Spatially resolved transcriptomics

1. INTRODUCTION However, most single-cell RNA sequencing (scRNA-seq)


approaches involve isolating cells from their original position,
Omics technologies, which provide a comprehensive, global meaning that spatial information is lost during transcriptome
assessment of a set of molecules,1 offer a novel approach for profiling. This omission makes it impossible to examine how
the study of biological activity in life-sciences research. In recent a given cellular state interacts with neighboring cells or the
years, the omics field has advanced considerably due to devel- surrounding extracellular matrix.2,5,6 To make up for this defi-
opments in the high-throughput analysis of biological molecules. cit, a new technology known as spatially resolved transcrip-
Transcriptomic technologies in particular have provided rich tomics (SRT) aims to determine gene expression profiles while
insights into cellular characteristics, and measure the complete set preserving information about the spatial context of the tissue.
of RNA transcripts produced by the genome under specific cir- This technique is of great significance, since identifying the
cumstances. Indeed, characterizing the transcriptome of individ- specific spatial location and organization of different cell types
ual cells is fundamental to improve our understanding of complex in multicellular tissues or organs is the foundation for research
biological systems. Over the last decade, the rapid development into cellular biological functions. Transcriptomics that is
of single-cell sequencing technology has greatly accelerated bio- accompanied by spatial information will undoubtedly enable
medical research, allowing scientists to overcome the major chal- us to better understand the organization of cells and tissues,
lenge of heterogeneity within biological samples and leading to and how this organization influences biological function.7 The
the publication of a series of single-cell transcriptome atlases.2–4 application of SRT has also opened up new areas of spatially
resolved omics, including genomic, transcriptomic, proteomic
and potentially other omic data with retained positional infor-
* Address correspondence: Hui Cheng, Tao Cheng, 288 Nanjing Road, Tianjin,
mation. The ability of SRT to change the way we understand
China. E-mail address: chenghui@ihcams.ac.cn (H. Cheng); chengtao@ihcams.
ac.cn (T. Cheng).
complex tissues earned it the title of Method of the Year 2020
in Nature Methods.8 In this review, we will first summarize
Conflict of interest: The authors declare that they have no conflict of interest.
and compare the major methods of SRT (Table 1), highlight
This work was supported by grants from the Ministry of Science and Technology some current applications of SRT, and finally discuss our per-
of China (2021YFA1100900 and 2020YFE0203000), the National Natural Science spectives on the potential future applications of this method.
Foundation of China (81730006, 81922002, 81861148029, and 81870086),
CAMS Innovation Fund for Medical Sciences (2021-I2M-1-040 and 2021-I2M-1-
019), Haihe Laboratory of Cell Ecosystem Innovation Fund (HH22KYZX0016) and
Distinguished Young Scholars of Tianjin (19JCJQJC63400). 2. METHODS AND ADVANCES
Blood Science (2023) 5, 1–14 The core, underlying principle of SRT, involves the acquisition
Received June 28, 2022; Accepted October 19, 2022. of gene expression profiles while retaining spatial information.
Broadly speaking, SRT methods can be classified into 3 groups:
http://dx.doi.org/10.1097/BS9.0000000000000141
imaging-based methods, sequencing-based methods, and in
Copyright © 2022 The Authors. Published by Wolters Kluwer Health Inc., on situ sequencing methods. Imaging-based methods are carried
behalf of the Chinese Medical Association (CMA) and Institute of Hematology,
out using a microscope, and the transcriptomes are read out
Chinese Academy of Medical Sciences & Peking Union Medical College
(IHCAMS). This is an open access article distributed under the Creative
through fluorescence in situ hybridization (FISH). Sequencing-
Commons Attribution License 4.0 (CCBY), which permits unrestricted use, based methods are performed using next-generation sequenc-
distribution, and reproduction in any medium, provided the original work is ing technology, and RNA with spatial information is captured
properly cited. and subsequently sequenced. In situ sequencing methods are
www.blood-science.org  1
Duan et al

Table 1
Major SRT methods.

Major methods of spatially resolved transcriptomics


Category Method Principle Strengths Limitations
smFISH smFISH In situ hybridization via complementary base High sensitivity Low throughput
pairing Subcellular resolution
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

Quantitative
Without amplification bias
Multiplexed FISH SRM Optical SRM and combinatorial labeling Improved throughput of smFISH Low throughput
ExFISH Physical magnification High-resolution imaging of RNA structure and Specific materials needed
location
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

seqFISH Sequential barcoding scheme Dramatically improved throughput Large number of designed probes needed
Optical crowding
Long workflow
MERFISH Error-robust encoding schemes achieved by Further improved throughput Large number of designed probes needed
introducing a certain Hamming distance Error detection and correction Optical crowding
Long workflow
corrFISH Cross-correlating the images from 2 of out of Decode high copy number RNAs Large number of designed probes needed
a total of 3 rounds of hybridization Long workflow
seqFISH+ Using secondary probes to dilute target Diluting the density of mRNA Large number of designed probes needed
transcripts Long workflow
Sequencing-based tomo-seq Cryosectioning of tissue and performing RNA-
Genome-wide RNA tomography of tissues or Often results in 1D data along one axis
methods that do not seq on individual thin sections organs Often only useful for elongated samples
validate cell types Slide-seq DNA-barcoded beads surface Near-cellular spatial resolution No tissue imaging
Slide-seqV2 High throughput
sci-Space Labeling nuclei using unmodified DNA oligos Single-cell resolution Imaging restricted to nuclei
on glass slides Large capture area
Stereo-seq DNA nanoball (DNB) barcoded solid-phase Nano-level resolution (Subcellular) Imaging restricted to nuclei
RNA capture Large capture area
Sequencing-based LCM-seq Applying laser energy to remove the cells of Speed, precision, and versatility Low throughput
methods that interest under microscopy Suitable for a small number of cells Damages adjacent cells
validate cell types RNA contamination
Geo-seq Combination of LCM and scRNA-seq Speed and precision Low throughput
Suitable for a small number of cells Damages adjacent cells
RNA contamination
NICHE-seq Cells sorted to scRNA-seq after Single-cell level RNA-seq Low throughput
fluorescent markers photoactivated by Reliant on transgenic mice
2-photon laser scanning microscopy Not suitable for clinical samples
ST Barcoded solid-phase RNA capture Combining histological staining Low spatial resolution
10X Visium Easy to perform Limited capture area
Requires no special instruments
Available commercial kit
HDST Barcoded solid-phase RNA capture Improved spatial resolution Limited capture area
DBiT-seq Microfluidic-based method to deliver Co-mapping of mRNAs and proteins Limited capture area
barcodes to the surface of a tissue slide Spatial resolution needs further
improvement
Seq-Scope Barcoded solid-phase RNA capture Nano-level resolution (Subcellular) Needs more research for demonstration
Large capture area
Sequencing-based TIVA Photoactivatable mRNA capture in live cells Compatible with live and intact tissue, mRNA Low throughput
methods in vivo and tissues captured with precise spatial resolution,
with little bias from RNA contamination or
experimentally-related injury
In situ sequencing FISSEQ Based on padlock probes and RCA, fixing the Preserve the tissue architecture for RNA Specific instruments required
methods cDNA fragments to the cellular protein matrix localization studies, localize subcellular RNA Random priming is inefficient and may
by amine cross-linker transcriptome-wide introduce bias
Large number of designed probes
needed
STARmap Based on padlock probes and RCA, 3D volumes of intact tissue, no intrinsic limit to Large number of designed probes needed
integrating hydrogel-tissue chemistry, the number of genes Workflow is complicated
targeted signal amplification, and In situ
SEDAL sequencing
Methods for FFPE Smart-3SEQ Combination of LCM and Smart-3SEQ Suitable for small samples or those whose RNA Needs more research for demonstration
tissue is degraded
Visium FFPE Probes capture RNA after tissue For clinical samples from biobanks, Needs more research for demonstration
decrosslinked immunological staining can be performed
corrFISH = correlation FISH, FFPE = formalin-fixed paraffin-embedded, FISH = fluorescence in situ hybridization, HDST = high-definition spatial transcriptomics, LCM = laser-capture microdissection,
MERFISH = multiplexed error-robust FISH, scRNA-seq = single-cell RNA-sequencing, smFISH = single-molecule FISH, seqFISH = sequential barcoded FISH, SRM = super-resolution microscopy, ST =
spatial transcriptomics, TIVA= transcriptome in vivo analysis, DBiT-seq =deterministic barcoding in tissue for spatial omics sequencing, ExFISH=expansion microscopy of fluorescence in situ hybridization,
FISSEQ=fluorescent in situ RNA sequencing, Geo-seq=geographical position sequencing, RCA= rolling circle amplification..

2 www.blood-science.org
 Chin Assoc of Blood Sci

the combination of imaging- and sequencing-based methods. However, N rounds of hybridizations bring not only exponentially
These methods have their intrinsic strengths and limitations, increased throughput, but also exponentially increased detection
with differences in target coverage, resolution, and throughput. error rates. When N increases, the error rates increase rapidly, and
Like all methods, they must be appropriately matched to the the majority of RNA molecules are likely to be misidentified after
biological question at hand. 16 rounds of hybridization.18 Therefore, multiplexed error-robust
FISH (MERFISH) method was designed. MERFISH is based on
combinatorial labeling and sequential imaging, and uses error-ro-
2.1. Imaging-based methods bust encoding schemes to correct labeling and detection errors.
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

Spatial information about protein expression can be obtained The method introduces a certain Hamming distance to encode
from traditional immunohistochemistry (IHC) approaches, RNAs, and the minimum Hamming distance is 4 (HD4 code),
which are mediated by the specific binding reaction between meaning that at least 4 bits must be read incorrectly to change
antigens and antibodies. In a variation of the IHC method, one codeword into another. As a result, every single-bit error pro-
spatial information about gene expression can be obtained by duces a word that is uniquely close to a single codeword, allowing
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

identifying the mRNA in cells using in situ hybridization (ISH), such errors to be detected and corrected. In addition, MERFISH
which involves a hybridization reaction between labeled nucle- improves the multiplexity of transcripts that can be obtained in
otide probes and complementary targeting RNA molecules. The single cells to 100 to 1000 through the use of binary sequential
single-molecule FISH (smFISH) method is the gold standard for barcodes19,20 (Fig. 1C).
detecting transcripts, and efforts to develop multiplex FISH at 2.1.3. Optical crowding: the major challenge for transcrip-
the transcriptome level are underway. This would allow FISH to
answer more complex biological questions. tome-level profiling Although seqFISH makes scaling to the
level of the genome theoretically possible, transcriptome-level
profiling in cells is hindered by optical crowding due to the
2.1.1. smFISH: the gold standard for transcript detec- high density of global transcripts, even when combined with
SRM. Some progress has been made to improve the resolu-
tion smFISH combines quantitative FISH with digital imaging tion of microscopy, with expansion microscopy (ExM) using
microscopy to sensitively detect single RNA molecules using a swellable polyelectrolyte gel that can expand physically to
probes. To amplify the signals, probes are often labeled with magnify the sample. RNAs are covalently attached to the gel,
multi-fluorochromes per molecule.9 However, the disadvantages, a small molecule linker, and FISH (ExFISH) is performed in
which include difficulty in acquiring heavily labeled oligonucle- cultured cells and intact tissues with high yield and specific-
otides, the easy loss of coupling fluorophores and self-quench- ity.21 Another approach called correlation FISH (corrFISH) was
ing, highly variable signals associated with multi-fluorophores used in seqFISH to read out individual RNA species and bar-
limit the system to a narrow range of applications. To overcome codes. The principle is simple: RNA species are decoded such
these drawbacks, singly labeled probes, which are easier to gen- that each RNA species appears in only 2 out of a total of 3
erate and purify, were introduced to accurately detect individual rounds of hybridization. By cross-correlating the images from
mRNA molecules with uniform signals10,11 (Fig. 1A). the 2 rounds of hybridization, only the specific RNA species will
smFISH can not only visualize the transcripts in isolated cells generate a positive correlation. Although some algorithms have
or in situ tissues at a high resolution and with high hybridiza- been adapted due to the need for a high degree of temporal reso-
tion efficiency, but also provides information about the distri- lution, corrFISH still represents a robust method to decode high
bution of subcellular RNA within cells. This feature means that copy number RNAs in highly multiplexed seqFISH experiments,
extra analyses concerning subcellular mRNA localization and using conventional fluorescence microscopy.22
its functional stages can be achieved, as mRNA localization is Another way to address the issue of molecular crowding is
currently believed to contribute to the localized regulation of to dilute the density of the mRNA. Thus, seqFISH+ labels the
gene expression.12–15 In addition, smFISH can precisely measure secondary probes with fluorophores rather than the primary
the copy numbers of specific RNAs without amplification bias probes, which provide target sites for the secondary probes.
and natural fluctuations in gene expression can be quantitatively The primary probes divide the transcript set into 20 subsets,
measured, permitting the elucidation of the mechanisms that reg- and the secondary probes, which are labeled with three col-
ulate gene expression fluctuations and their role in a variety of ors, ultimately make it possible to visualize one-sixtieth of the
biological processes. These features make smFISH a robust tool, transcripts per image in each round of hybridization (Fig. 1D).
and it is the standard method used for detecting transcripts. seqFISH+ achieves transcriptome-level profiling not only for
cultured cells but also in tissues, so that unbiased identification
2.1.2. Multiplexed FISH: improving target coverage Many of cells alongside their spatial organization can be realized.23
biological questions require transcription information about mul-
tiple genes, so efforts have been made to address the challenge
of the limited number of RNA species that be simultaneously 2.2. Sequencing-based methods
detected in cells using FISH. Super-resolution microscopy (SRM) With the development of genetic techniques, next-generation
can resolve a large number of mRNAs in single cells by labeling sequencing technology has become a powerful tool for tran-
mRNAs with unique combinations of fluorophores. Although this scriptome analysis. scRNA-seq can precisely characterize cell
expanded the scalability of FISH from a few genes to about 30, it types and states at the molecular level, without bias, based on
was hard to make further progress due to the limited number of flu- global gene expression profiles.24 Generally speaking, sequenc-
orophores.16 Later, a sequential barcoding scheme was introduced ing-based methods are performed to capture or barcode the
to multiplex different mRNAs, called sequential barcoded FISH RNA before reverse transcription to ensure each transcript can
(seqFISH).17 Here, the mRNA is barcoded by sequential rounds of be mapped to its original spatial spot, or are used to gain gene
hybridization, imaging, and probe stripping. The number of RNA expression information from cells in a certain spatial context.
species scales as FN, where F is the number of fluorophores and N RNA-seq can be conducted either ex situ or in situ.
is the number of rounds of hybridization. Thus, the scalability of
seqFISH can be dramatically improved by increasing the number
of rounds of hybridization, despite the limited number of fluo- 2.2.1. Sequencing-based methods that do not validate
rophores. In this manner, the entire human transcriptome can be cell types Some methods do not provide the spatial resolu-
covered, including both mRNAs and non-coding RNAs (Fig. 1B). tion of microscopy-based techniques as they do not predict or

www.blood-science.org  3
Duan et al
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

Figure 1. Schematics of the main imaging-based methods. (A) In smFISH, fluorochrome-labeled probes detect individual mRNA molecules through in situ
hybridization. (B) In seqFISH, sequential barcoding: in each round of hybridization, a set of probes targeting different genes are hybridized on each transcript,
imaged, and then stripped with DNAse I. The same probe sequences are used in different rounds of hybridization, but probes are coupled to different fluoro-
phores. By decoding the temporal color sequences, the original genes can be identified. (C) In MERFISH, HD4 code is introduced by encoding probes contain-
ing a central RNA-targeting region flanked by 2 readout sequences. Encoding probes convert the RNA into a unique combination of readout sequences. (D) In
seqFISH+, I–IV sequences on the primary probes correspond to 4 rounds of barcoding, and the fourth round is used for error correction. Only one-twentieth
of the total genes in each fluorescent channel are labeled by readout probes in each hybridization readout round, lowering the density of transcripts in each
image. HD4 = minimum Hamming distance is 4, MERFISH = multiplexed error-robust FISH, seqFISH = seqFISH = sequential barcoded FISH, smFISH = sin-
gle-molecule FISH.

validate cell types using IHC or ISH. Examples of such methods of the body. For this reason, tomo-seq is best suited for elon-
include tomo-seq and Slide-seq.25,26 Tomo-seq is a serial micro- gated samples. Cells with complex organizations in the same
tomy-based sequencing method, which cryosections the tissue individual sections cannot be analyzed well, and often several
of interest and performs RNA-seq on individual thin sections identical samples are required to perform cryosectioning in dif-
(Fig. 2A). RNA extraction is performed for each individual ferent directions along the main body axes to achieve 2- and
section and the RNA is barcoded with section-specific primers. 3-dimensional resolution.27
This approach can provide genome-wide RNA tomography of Unlike tomo-seq, Slide-seq is a method for transferring RNA
tissues or organs, but it is often only possible to cryosection tis- from tissue sections onto a surface covered in DNA-barcoded
sue in 1 direction, resulting in 1-dimensional data along 1 axis beads with known positions, allowing the locations of the RNA

4 www.blood-science.org
 Chin Assoc of Blood Sci
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

Figure 2. Schematics of the main sequencing-based methods. (A) In tomo-seq, specimens are cryosectioned, and sections are collected in individual micro-
tubes for spatially resolved transcriptomic analysis. (B) In LCM, an infrared laser melts the thermolabile polymer on a tissue section on a glass slide in the vicinity
of the laser pulse, resulting in the removal of polymer-cell composite from the tissue. A UV laser can cut away cells of interest or ablate unwanted tissue, leaving
cells of interest intact on the slide. (C) In NICHE-seq, tissue expressing a PA-GFP can be activated by 2-photon irradiation, allowing precise in situ labeling.
Activated cells are sorted to perform MARS-seq. (D) In 10X Genomics’ Visium, tissue sections are placed on a barcoded glass slide containing 4 capture areas,
each with around 5000 spatial spots. After HE staining and imaging, tissue permeabilization releases the mRNA and it is captured by spatial probes. LCM =
laser-capture microdissection, PA-GFP = photoactivatable green fluorescent protein, MARS-seq =massively parallel scRNA-seq, UV=ultraviolet, HE staining =
hematoxylin-eosin staining.

to be inferred by sequencing. It provides a scalable method for The hashed oligos spotted onto the glass slides are transferred
obtaining spatially resolved gene expression data at resolutions onto permeabilized nuclei (not intact cells) in freshly frozen tissue
comparable to the sizes of individual cells.26 Slide-seqV2 is an opti- sections. As the nuclei are stained with 4',6-diamidino-2-phenylin-
mized version of Slide-seq with improvements in library genera- dole (DAPI) and the spatial coordinates are consequently imaged
tion, bead synthesis, and array indexing to reach an RNA capture during oligo transfer, sci-Space can retain a single-cell resolution.29
efficiency ~10-fold greater than Slide-seq.28 A new method, called Obviously, the main limitation of all these methods is that the cell
sci-Space, adapts the low-cost sci-Plex procedure for labeling or types are purely identified by RNA-seq datasets, which lacks the
“hashing” cell nuclei using unmodified DNA oligos to glass slides. cell-validation process that microscopy-based techniques provide.

www.blood-science.org  5
Duan et al

2.2.2. Sequencing-based methods that validate cell tissue slide, so that proteomics can be integrated with transcrip-
types Although single-cell RNA-seq has the power to define tomics. The capacity for integration is of great significance for
cell types or populations based on gene expression programs, multi-omics research.
microscopy-based techniques such as IHC or ISH—which val-
idate candidate genes with high cell type specificity—remain
the gold standard to define cell type. Thus, a series of sequenc- 2.2.3. Sequencing-based methods for use in vivo Current
ing-based methods combined with imaging have been developed methods that capture RNA, such as LCM, cannot isolate mRNA
to aid the study of solid cell types. from individual in vivo cells without damaging adjacent tissue,
making it difficult to assess the influence of the microenviron-
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

Laser-capture microdissection (LCM) is an optional method


to obtain targeted cell subgroups or even single cells quickly ment on the transcriptome. To overcome this problem, a method
and precisely under the microscope. LCM directly harvests cells was engineered to extract mRNA from live cells or tissues at the
of interest or isolates specific cells by cutting away unwanted spatial resolution of a single cell. This method is called transcrip-
cells via infrared capture systems or ultraviolet cutting systems30 tome in vivo analysis (TIVA); it uses a photoactivatable bioti-
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

(Fig. 2B). Histological staining or rapid antibody staining are nylated tag, known as a TIVA-tag, that can penetrate the cell
performed on the tissues before LCM so that information about membrane by virtue of a cell-penetrating peptide. Once inside
the types of cells captured is validated. The key advantages of the cell, photoactivation is used to cleave the linkers between
LCM are that the positional information of cells is maintained the poly(U) sequence and two poly(A) stretches to enable the
and tissues are not dissected; in this way, subsequent analysis by poly(U) sequence to bind the poly-A tail of mRNAs (Fig. 3B).
genomics, transcriptomics, or proteomics can be performed on With this method, mRNA can be noninvasively captured within
the LCM-captured cells.31–33 The extracted nucleic acids can be live cells and intact tissues, so it provides a useful tool to explore
amplified when the captured regions of interest are small, over- the transcriptomes of single cells in the context of their natu-
coming the requirement for relatively large numbers of cells. ral microenvironment. However, the method is low throughput
This advantage makes it possible to use LCM at the single-cell with regard to the numbers of cells that can be analyzed.
level, meaning that it is especially useful for scarce tissues and 2.2.4. Enhanced resolution and a larger area permit the
rare cell types. Geo-seq is the combination of LCM and scRNA- acquisition of more detail Solid RNA-barcoding is favor-
seq technology.33 It is also practical to combine LCM with able in terms of its untargeted features and high coverage com-
Smart-seq2, a high-throughput scRNA-seq technology with pared with imaging-based methods. Efforts have been made to
improved sensitivity, accuracy, and full-length coverage across improve the spatial resolution and capture area of this method,
transcripts, making LCM plus Smart-seq2 a promising prospect as a higher resolution means the precise location of the tran-
for SRT.31,34,35 However, the main obstacles to the wide appli- scripts within the tissues, cells or even subcellular organelles can
cation of LCM SRT are its low throughput and potential RNA be determined.
contamination from other cells, as well as the inevitable damage ST can achieve a resolution of only 100 μm (which is not at
to adjacent tissue. This means that it is difficult to study the the single-cell level), which means that the 100 μm spatial spots,
interplay between individual cells and the microenvironment. with a center-to-center distance of 200 μm, typically cover 5 to
One method with similarities to LCM coupled with Smart- 100 cells each, depending on tissue type and region.37 Visium
seq2 is NICHE-seq, which couples with scRNA-seq. This tech- technology from 10X Genomics has improved the ST method
nique combines photoactivatable fluorescent markers, 2-photon by reducing the diameter of the capture area from 100 to 55 μm,
laser scanning microscopy and flow cytometry-based fluores- increasing the capture area to 42.25 mm2 (6.5 mm × 6.5 mm),
cence-activated cell sorting (FACS), and is coupled to massively and decreasing workflow duration (Fig. 2D). Nevertheless, the
parallel scRNA-seq (Fig. 2C). NICHE-seq requires transgenic spatial resolution of 10X Genomics’ Visium is still insufficient to
mice that ubiquitously express a photoactivatable green fluo- resolve single cells in most samples.
rescent protein (PA-GFP). Two-photon irradiation is used to Slide-seq and DBiT-seq both have improved spatial resolution
activate PA-GFP and in situ labeling. After tissue dissociation, to 10 μm,26,38 but this is still a near-cellular resolution and cannot
GFP-labeled cells can be sorted to perform scRNA-seq; thus, be described as single-cell resolution. High-definition spatial tran-
this method combines information about the cell’s transcrip- scriptomics (HDST), an optimized version of ST, was designed
tional state and spatial information.36 As this method requires to improve ST resolution to the single-cell level. In this method,
transgenic mice expressing GFP, however, it is not suitable for hundreds of thousands of barcodes are randomly deposited onto
clinical samples. a slide at 2 μm resolution, and their positions are decoded by
Another method based on barcoded glass slides, called spa- sequential hybridization and error-correcting strategy.39 One
tial transcriptomics (ST), also validates cell types by histolog- method, called spatiotemporal enhanced resolution omics-se-
ical staining and imaging.37 RNA is spatially captured from quencing (Stereo-Seq), involves the deposition of a DNA nanoball
tissue sections and barcoded via a microarray on a glass slide. containing random barcoded sequences to form the chip. Each
Several primers with unique barcodes are placed on the micro- spot is approximately 220 nm in diameter, with a center-to-cen-
scopic glass slide to create a microarray, and a section of tissue ter distance of 500 nm.40 Another method, known as Seq-Scope,
is placed on top of the microarray surface. This tissue is then has a center-to-center resolution of 0.5 to 0.8 μm (0.6 μm on
fixed, then stained and imaged, before being permeabilized to average).41 The nano-level distances of these methods are far less
release RNA within the cells. In this way, transcripts from the than the diameter of single cells, giving them the high resolution
tissue are allowed to meet with the immobilized cDNA synthesis needed to determine the subcellular location of transcripts.
primers in a reverse transcription reaction. Consequently, the
cDNA library is sequenced and the data are visualized together
with a high-resolution histological image of the tissue section. 2.3. In situ sequencing methods
Each slide consists of about 1000 spots, and ST is both easy to
perform and requires no special instruments. The method has The combination of imaging and sequencing processes typ-
been acquired and optimized by 10x Genomics and a commer- ically involves in situ sequencing methods. In situ sequencing
cial kit is available (Fig. 2D). methods sequence cDNA amplicons based on padlock probes
DBiT-seq is a microfluidic-based method that delivers bar- and rolling circle amplification.42,43 The 2 ends of the padlock
codes to the surface of a tissue slide to allow for spatial omic probe can be ligated to form a circle-like structure upon hybrid-
sequencing. This method enables the co-mapping of mRNAs ization, which then can be amplified using an isothermal DNA
and proteins on a slide containing formaldehyde-fixed tissue38 polymerase. Amplification causes copies of the padlock probe
(Fig. 3A). Immunofluorescence can be performed on the same sequence to accumulate and form cDNA nanoballs. An amplified
6 www.blood-science.org
 Chin Assoc of Blood Sci
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

Figure 3. Schematics of DBiT-seq, TIVA, and STAR map. (A) In DBiT-seq, parallel microfluidic channels are used to deliver DNA barcodes to the surface of a
tissue slide and yield a 2D mosaic of tissue pixels. ADTs recognize a panel of proteins of interest, co-mapping mRNAs and proteins. (B) In TIVA, caged TIVA-
tags are loaded into cells or tissue by virtue of a disulfide-linked CPP. Selective photoactivation of the TIVA-tags in the desired cell or cells using a laser uncages
the TIVA-tags, and the exposed poly(U) can anneal to the poly-A tail of cellular mRNA. (C) In STAR map, only when both probes (SNAIL probes) hybridize to
the same RNA molecule can the padlock probe be circularized and rolling-circle-amplified to generate a DNA nanoball. The amplicons with an acrylic acid
N-hydroxysuccinimide moiety modification are copolymerized with acrylamide to embed within a hydrogel network. Gene identifiers are sequenced by in situ
SEDAL sequencing. ADTs = antibody-derived DNA tags, CPP = cell-penetrating peptide, STAR = spatially-resolved transcript amplicon readout mapping, TIVA=
transcriptome in vivo analysis, DBiT-seq =deterministic barcoding in tissue for spatial omics sequencing, SEDAL=sequencing with error-reduction by dynamic
annealing and ligation.

signal can be obtained by visualizing the cDNA nanoballs using FISSEQ, a highly multiplexed subcellular in situ RNA
fluorescently labeled detection oligonucleotides, which are com- sequencing method, uses random hexamer-primed reverse
plementary to the padlock probe. transcription in the presence of aminoallyl dUTP. Aminoallyl
www.blood-science.org  7
Duan et al

dUTP then fixes the cDNA fragments to the cellular protein commercially available system, called Visium FFPE, that enables
matrix by a non-reversible amine cross-linker.44 The templates SRT to be performed on samples from a biobank. Some stud-
are circularized after degrading the RNA, and subsequently ies have already successfully used 10x Visium FFPE on FFPE
amplified using rolling circle amplification primers comple- tissue.48
mentary to the adapter sequence introduced by RT. The ampl-
icons in the cells are then ready for sequencing-by-ligation
(SOLiD sequencing) at room temperature, and imaging on a 3. DATA ANALYSIS IN SRT
confocal microscope. To control the signal density of FISSEQ, The additional dimension of spatial information brings not
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

a partition sequencing strategy was devised using pre-extended only a novel perspective on the transcriptome, but also signif-
sequencing primers.44,45 With 1 base extended, one can ran- icant challenges for data analysis due to increased data vol-
domly sample amplicons at 1/4th of the original density, and ume and complexity. Generally speaking, strategies for SRT
with 2 bases extended sampling at 1/16th of the density is pos- data analysis include computational approaches specifically
sible. One notable highlight of FISSEQ is that it preserves the designed for SRT and integration methods (with bulk or sin-
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

tissue architecture for RNA localization studies, and localizes gle-cell RNA-seq data). Different analytical approaches have
subcellular RNA transcriptome-wide. However, the random been developed for localized gene expression pattern identifi-
priming component is inefficient and may introduce bias via cation, spatial decomposition, gene imputation, and cell–cell
uneven binding efficiency, which is due to differences in base communication.49
composition. Gene expression and spatial location are both of great
While SRT can be performed on tissues or sections on a slide, relevance to biological functions. By analyzing the relation-
applying in situ sequencing methods to 3-dimensional volumes ship between gene expression and spatial location, some
of intact tissue is not easy. To achieve this goal, a method known approaches, including Trendsceek,50 SpatialDE,51 SPARK,52
as spatially resolved transcript amplicon readout mapping SPARK-X,53 sepal,54 SpaGCN,55 and GLISS, are designed to
(STARmap) integrates hydrogel-tissue chemistry, targeted signal identify localized gene expression patterns and spatially vari-
amplification and in situ sequencing.46 The method bypasses the able genes (SVGs) based on different algorithms. Another way
reverse transcription step and uses the SNAIL approach instead, to discover SVGs is spatial clustering. Unlike standard clus-
which achieves specific amplification of nucleic acids via intra- tering methods for scRNA-seq, spatial clustering is for the
molecular ligation using a pair of custom primer and padlock capture locations. Methods like stLearn, MULTILAYER,56
probes. Only when both probes hybridize to the same RNA BayesSpace,57 SC-MEB,58 and STAGATE can be applied for
molecule can the padlock probe be circularized and rolling-cir- spatial clustering. As most of the sequencing-based methods
cle-amplified to generate a DNA nanoball. The cDNA ampli- do not guarantee that a single capture spot contains RNA
cons are modified and copolymerized in situ to embed within a from only 1 cell, a capture location is often a mixture of mul-
hydrogel network. In situ SEDAL sequencing was devised specif- tiple cell types. Thus, some spatial decomposition algorithms,
ically for STARmap, to sequence 5-base barcoded gene-specific such as NNLS, spatialDWLS,59 SPOTlight,60 RCTD,61 DSTG,
identifier segments in SNAIL probes over 6 rounds of hybridiza- Tangram, and Cell2location attempt to infer the proportions
tion (Fig. 3C). The highlight of STARmap lies in its ability to be of cell types in each spot, or score a spot for how strongly
used in 3-dimensional in situ transcriptomics, which is undoubt- it corresponds to a single cellular subtype based on bulk or
edly a state-of-the-art SRT technique and serves to deepen our single-cell RNA-seq data. However, certain SRT methods,
understanding of anatomy and transcriptomics. In parallel, particularly those that are imaging-based, cannot provide
there is no intrinsic limit to the number of genes or RNA spe- deep transcriptomic information due to limited gene cover-
cies that can be simultaneously and quantitatively accessed with age. Gene imputation provides a chance to impute the missing
STARmap, since it can be adapted to longer sequencing lengths genes from scRNA-seq data in order to improve the quality
or higher gene numbers. of SRT data. Methods that include gene imputation functions
In conclusion, ISH is highly sensitive, and can detect tran- include Tangram,62 gimVI, Harmony,63 LIGER,64 Seurat,65
scripts at a subcellular resolution. Many powerful strategies SpaGE,66 and stPlus.67 In addition, the reconstruction of spa-
have been designed to improve the target coverage of ISH tial information for scRNA-seq data can be achieved by map-
to the transcriptome level and tackle the problem of opti- ping, which leverages a small set of landmark genes to map
cal crowding. However, such methods are typically limited the single cell back to its context. Methods that incorporate a
by long image acquisition times and a complex probe-de- mapping function include Harmony, LIGER, Seurat, SpaGE,
signing process. While next-generation sequencing technol- DEEPsc,68 DistMap, SpaOTsc,69 novoSpaRc,70 and CSOmap.71
ogy enables high-throughput transcriptomic profiling, many In addition, cell–cell interactions can be analyzed using some
sequencing-based methods are much easier to perform once standard algorithms, based on the ligand–receptor interaction
suitable experimental platforms have been established. As pairs from scRNA-seq data and a database of known ligand–
spatially barcoded beads or spots can be arrayed more and receptor interactions. However, the loss of spatial information
more densely, some solid-barcoded methods can reach a cellu- in scRNA-seq data may lead to inaccurate detection of ligand–
lar or subcellular resolution, and spatial information is more receptor interactions, as cellular cross-talk is often spatially
precisely retained. To some extent, in situ sequencing methods restricted. Therefore, SRT data might potentially evaluate the
are a combination of imaging- and sequencing-based methods, reliability of the ligand–receptor interactions computed from
sequencing gene identifiers in the designed primers via FISH. scRNA-seq. In this manner, integrating scRNA-seq with SRT
Although in situ sequencing methods are high throughput, the data may accurately reveal specific cell subpopulations and
complexity of designing targeted probes cannot be ignored their interactions. Approaches to study cell–cell interactions
(Table 1). include SVCA,72 GCNG, NCEM, MISTy, stLearn, Squidpy,
Most of the in situ-based methods described above rely on the novoSpaRc, SpaOTsc, and DEEPsc. Among these meth-
use of fresh tissues. However, fresh clinical material is often dif- ods, comprehensive benchmark studies are needed to help
ficult to obtain for research purposes. To remove the barriers to users select methods that best fit their data and hypotheses.
clinical research, some methods have been designed to use for- According to a study benchmarking 16 integration methods,
malin-fixed paraffin-embedded (FFPE) tissue, in which the RNA Tangram, gimVI, and SpaGE outperformed other integration
is degraded. Smart-3SEQ enables large gene expression profiling methods when predicting the spatial distribution of RNA
experiments to be conducted on even small amounts of total transcripts, while Cell2location,73 SpatialDWLS, and RCTD
RNA, and effectively characterizes small samples extracted by were the top-performing methods for the cell type deconvo-
LCM from FFPE tissue.47 In addition, 10X Genomics markets a lution of spots.74
8 www.blood-science.org
 Chin Assoc of Blood Sci

In conclusion, SRT and scRNA-seq data complement each with varying genetic alterations and the interactions between
other, since SRT methods cannot always maintain single-cell res- tumor cells and the tumor microenvironment presents a compli-
olution with the same depth and whole transcriptome coverage cated situation that influences disease development.82 Profiling
of scRNA-seq. The integration of scRNA-seq and SRT data could the spatial heterogeneity of tumors and the tumor microen-
improve cell type annotation, cell clustering, spatial decompo- vironment will provide insights into tumor progression and
sition, gene imputation, and spatial location reconstruction. A responses to treatment, and will therefore assist in the diag-
scRNA-seq database could form a solid foundation for SRT data nosis and treatment of this condition. ST technology has been
analysis. However, the proper handling of spatial information used to identify intertumoral and intratumoral heterogeneity in
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

obtained from SRT data remains a challenge in terms of data multiple types of cancer. Furthermore, changes in gene expres-
processing, and the establishment of proper protocols will be an sion during prostate cancer and cutaneous malignant mela-
essential step to reach the full potential of these methods. noma progression have revealed a detailed landscape of tumor
progression and metastases.83,84 Several features of the tumor
microenvironment in primary liver cancer, including stromal
4. APPLICATIONS FOR SRT
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

and immune cell distribution, tumor cluster interaction, can-


At present, SRT technologies are widely utilized in basic and cer stem cell-niche diversity and tertiary lymphoid structure
clinical research, especially in neuroscience, cancer biology, and composition have been characterized.85 Coupling SRT with
developmental biology. However, there are likely further appli- other technologies also improves its capacity to characterize
cations for SRT that are yet to be fully utilized, including in the tumors: for example, when SRT was combined with scRNA-
study of hematopoeisis. We reviewed the potential applications seq, tissue architecture and interactions between cell subpopu-
in this field in Figure 4. lations were revealed in pancreatic ductal adenocarcinomas.86
A tumor-specific keratinocyte population, immune infiltrates,
and heterogeneity at tumor leading edges have also been iden-
4.1. Neuroscience: unraveling the organization of brain tified in human squamous cell carcinoma using multimodal
cell types and functions analysis, which included scRNA-seq and ST.87 The heteroge-
The diversity of brain cell types and the complex organization neity of breast cancer has also been characterized, and several
of the brain have long been major challenges to our understand- diagnostic and prognostic markers proposed.88,89 Additionally,
ing of brain functions during physiological and pathological by integrating spatial gene expression and HE-stained images
conditions.75 A census of brain cell types and their functions will of breast tumor, a deep learning algorithm was developed to
pave the way for systematic research and, potentially, even novel predict the spatially resolved transcriptome of a tissue from
clinical treatments. SRT has advantages over scRNA-seq due to the images, enabling image-based screening for molecular bio-
the retained anatomical and spatial contexts, and various spa- markers.90 To guide clinical treatment, biomarkers associated
tially resolved transcriptomic methods have been applied to map with beneficial PD-1 checkpoint blockade in non-small-cell
cell types in neuroscience. For example, an atlas of the mouse lung cancer were identified using digital spatial profiling.91
somatosensory cortex was built using osmFISH, a semiautomated The ST analysis of human bladder cancer also identified an
version of smFISH. Based on scRNA-seq data, the study identified N-Cadherin 2-expressing epithelial cell subpopulation which
31 clusters of cell types and 33 cell type-specific marker genes.76 could be used to predict therapeutic response, which could
Although multiplexing in osmFISH is relatively low, its sensitivity guide treatment decisions.92 In conclusion, these findings pro-
is higher than scRNA-seq and can provide single-cell-level spatial vide novel insights into the complex ecosystem of cancer. and
resolution for gene detection. Multiplexed FISH techniques, such have the potential to improve individualized cancer treatments
as MERFISH and seqFISH, are applied when more genes need and drug discovery.
to be analyzed. The multiplexing of 250 genes using seqFISH
identified distinct subregions of the hippocampus that contained
different combinations of cell types.18 In addition, MERFISH 4.3. Developmental biology: characterizing the
combined with measurements of immediate early gene expression temporal and spatial expression blueprint of embryonic
identified ~70 neuronal populations in the mouse hypothalamic development
preoptic area, and defined discrete cell populations activated by
specific social behaviors.77 STARmap has also been used to recon- Embryonic development is a complex process where dynamic
struct the 3D locations of cells in the murine primary visual cor- changes rapidly occur at the biomolecular level.93,94 Lineage
tex and medial prefrontal cortex; in this particular study, >1000 tracing and fate mapping are powerful tools in developmen-
genes were simultaneously mapped in mouse brain sections at a tal biology, providing a picture of the lineage hierarchy and
single-cell resolution.46 Sequencing-based methods such as Slide- linking the initial cell position to future fate.95 With the emer-
seq and 10X Genomics’ Visium have also been used to analyze gence of SRT, analysis of both the temporal and spatial aspects
the global transcriptome in both the mouse and human brain.26,37 of embryonic development can be performed, and a temporal
The application of spatially resolved transcriptomic methods and spatial expression blueprint is eagerly anticipated. In the
is highly likely to improve our understanding of neurological early embryo, gastrulation is an essential developmental event,
diseases. For example, SRT has already revealed the spatiotem- leading to the formation of gastruloids, which are aggregates
poral dynamics of molecular pathology in amyotrophic lateral of embryonic stem cells.96,97 Applying Geo-seq to the germ lay-
sclerosis.78 Furthermore, in a murine model of Alzheimer dis- ers from pre- to late-stage gastrulation enabled the molecular
ease, ST and in situ sequencing were used to better characterize genealogy of tissue lineages and the continuum of pluripotency
the dysregulated cellular network in the early and late phases of states in time and space to be revealed. Furthermore, the molec-
disease. The results of this study provided profound insights into ular determinants that drive lineage specification and tissue
the pathogenesis of Alzheimer disease, and also provided clues patterning were identified.98 Tomo-seq and scRNA-seq revealed
concerning potential therapeutic targets.79 genome-wide gene expression patterns in mouse gastruloids and
embryos, and identified various embryonic cell types that were
not previously known to be present in gastruloids. Based on
4.2. Cancer: profiling the spatial heterogeneity of tumors their results, the study’s authors proposed that somitogenesis
occurs in gastruloids.99 In addition, ISH has determined the gene
and the tumor microenvironment expression pattern in gastruloids generated from embryonic
Heterogeneity both within and between tumors is found in stem cells, to validate an in vitro model of early anteroposterior
most types of cancer.80,81 The expansion of tumor subclones organization during human development.100 Stereo-seq has also
www.blood-science.org  9
Duan et al
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

Figure 4. Applications for SRT. In neuroscience, SRT was used to unravel the organization of brain cell types and functions. In cancer biology, the spatial het-
erogeneity of tumors and the tumor microenvironment can be characterized. In developmental biology, SRT was used to characterize the temporal and spatial
expression blueprint of embryonic development. In hematology, most studies utilizing SRT concern embryonic hematopoiesis, and aim to clarify the distinct
waves of developmental hematopoiesis. AGM = aorta-gonad-mesonephros, HSCs = hematopoietic stem cells, SRT = spatially resolved transcriptomics.

reconstructed the spatially resolved developmental trajectories and definitive hematopoiesis.107,108 SRT technologies have been
of cell-fate transitions and molecular changes during zebrafish applied in developmental hematopoiesis to explore the genera-
embryogenesis.101 tion and expansion of HSCs, as well as the regulation of their
Some research has also focused on the formation of organs in niche. Geo-seq, combined with bulk and single-cell RNA-seq,
embryonic development. The large field-of-view and cellular res- was used to examine the caudal hematopoietic tissue of zebraf-
olution of Stereo-seq has been used to map the spatiotemporal ish (the counterpart of the mammalian fetal liver) and a detailed
transcriptomic dynamics during mouse organogenesis.102 ST was spatiotemporal transcriptome of hematopoietic stem and pro-
also used to study cardiac morphogenesis, and the spatiotempo- genitor cells (HSPCs) and niche during HSPCs expansion was
ral organ-wide gene expression and cell atlas of the developing characterized.109 Meanwhile, LCM-seq has been used in the
human heart was established during 3 developmental stages.103 human embryonic aorta-gonad-mesonephros (AGM) region to
Morphogenesis of the human intestine across time, location, identify secretory factors that promote human HSC develop-
and cellular compartments has been charted by both scRNA- ment.110 Another study combined with 10X Genomic’s Visium
seq and 10X Genomics’ Visum.104 The gene expression profile of and Stereo-seq identified novel HSC/MPP pocket-like units
fetal livers from 8 to 17 weeks post-conception in humans has (HSC PLUS) composed of niche cells and enriched with growth
also been illustrated using 10X Genomics’ Visium.105 factors; providing an essential resource for understanding HSC/
MPP development in the fetal liver.111 In addition, 10X Genomic’s
Visium was used to visualize HSC emergence in human embry-
4.4. Hematopoiesis: shedding light on the distinct onic tissues, including the liver, AGM, gut, and vitelline and
waves of developmental hematopoiesis, and making umbilical vessels, validating the generation of AGM-like defini-
tive HSPCs from human pluripotent stem cells.112
breakthroughs in adult bone marrow scRNA-seq technologies have been widely performed in the
Hematopoiesis is the process by which blood cells are con- adult hematopoietic system, and have confirmed the heteroge-
tinually replenished throughout the lifetime of an organism, neity of HSPCs.113 However, SRT is yet to be applied in this
by virtue of the self-renewal and differentiation of hematopoi- field. Profiling the SRT of adult bone marrow would be of great
etic stem cells (HSCs) and the regulation of the local micro- significance because this will provide an opportunity to study
environment, or niche.106 Distinct waves of hematopoiesis have the interplay between HSPCs and their niche under both nor-
been defined during embryogenesis, including both primitive mal conditions and stress. Most SRT methods require an intact
10 www.blood-science.org
 Chin Assoc of Blood Sci

slice of tissue, which may be challenging for adult bone marrow rapid membrane imaging methods, such as fluorescent probe-
due to the presence of calcified bone. Therefore, tissue prepa- based methods, have already been used to facilitate in situ
ration and cryosectioning need to be optimized. Furthermore, plasma membrane imaging of neurons and erythrocytes in the
bone marrow is composed of various hematopoietic cells and complex environment of the brain.119 However, optimization of
non-hematopoietic cells, including mesenchymal stromal cells, in situ plasma membrane imaging in combination with SRT still
pericytes, fibroblasts, and endothelial cells.114,115 Importantly, requires further work.
bone marrow hematopoietic cells are usually smaller than other
cell types, and the cells are usually distributed evenly in the bone
5.2. Integrating spatially resolved multi-omics to advance
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

marrow, lacking any apparent structure in terms of anatomi-


cal or functional division.116,117 These intrinsic features of bone our understanding of biological systems
marrow impede the experimental applications of SRT, as well
SRT is far more than just transcriptomics; rather, the tech-
as associated data processing. LCM-seq has been applied to
nique is empowered by the retained positional information
study adult bone marrow to reveal bone marrow niche organi-
provided by in situ methods. The locations of cells and the
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

zation, setting an excellent example for future research.118 RNA-


microenvironment play an important role in uncovering cell-to-
barcoding methods with improved resolution, such as Slide-seq,
cell and cell-to-extracellular matrix interactions, and the inte-
HDST and stereo-seq, are recommended for bone marrow, since
gration of functional and spatial information is of great benefit
the cellular or subcellular resolution is the key to compensating
in biological studies. Spatially resolved transcriptomic methods
for smaller cells and diverse cell types. Furthermore, different
provide several invaluable methods that can also be adapted to
types of HSPCs share high similarities in transcriptomics. Thus,
suit certain other in situ omics. We expect that SRT will inspire
we propose that SRT data can be properly analyzed only when
the development of other spatially resolved omics beyond
combined with scRNA-seq data. Of note, in the past decade,
transcriptomics, such as in situ epigenomics, epitranscriptom-
single-cell transcriptomic data of bone marrow hematopoietic
ics, proteomics, and metabolomics, which will help us better
cells and niche cells has been collected, serving as a supporting
understand different aspects of cell function. Epigenomics is a
resource for SRT.114,115
reliable tool to map chromatin accessibility dynamics and high-
er-order chromatin structure, and adding spatial information to
this would surely enable new levels of understanding of cell-
5. CONCLUSIONS AND FUTURE PERSPECTIVES
fate decisions, identity, and function.120,121 Epitranscriptomics
SRT technologies provide an opportunity to uncover the concerns a wide range of post-transcriptional RNA modifi-
molecular architecture of tissues. Methods that are more scal- cations, and spatial epitranscriptomics enables studies under
able, have a higher resolution and easier workflows are expected specific microenvironments, broadening our understanding of
to be developed, and more results produced by SRT should be the spatial regulation of gene expression.122 In parallel, metab-
published soon. For the remainder of this review, we anticipate olomics transcends genomics and proteomics, representing the
future developments that will unlock the full potential of SRT in most downstream metabolic stage. The metabolome is widely
both biological research and the clinic. accepted to constitute a dynamic and sensitive measure of phe-
notype, placing metabolomics at the forefront of biomarker
discovery.123 Several spatially resolved metabolomic technolo-
5.1. Realizing single-cell SRT gies have already been applied in several fields, such as tumor
The correct association of detected mRNAs and single cells is metabolism, allowing metabolites and metabolic enzymes to be
a great challenge for both imaging- and sequencing-based meth- directly discovered and studied in their native state.124–127
ods. For most sequencing-based methods, such as Slide-seq and On the other hand, cell states that are defined based on data
ST,26,35,37 the spatial information is retained in the form of dif- from one type of omics may not be accurate and comprehensive,
ferentially barcoded spots. Although some methods, including and cell states defined by different omics may not always be in
HDST, Stereo-seq and Seq-Scope, have achieved cellular or sub- accordance. We therefore expect the emergence of more spatially
cellular resolution,39–41 the spot boundaries do not correspond resolved multimodal technologies, both at the experimental
to the cell boundaries, meaning that transcripts captured in the level and in computational analysis, to be able to simultane-
same spot do not always come from a single cell. Furthermore, ously profile multiple data types in the same cell. For exam-
it cannot be guaranteed that the 10 μm-thin slices usually ple, DBiT-seq can capture both proteins and transcripts on the
obtained during tissue sampling will contain a single layer of same sequencing slide, so that proteomics and transcriptomics
cells. This means that the transcripts of all the cells on the Z-axis can be simultaneously performed on the same cells or tissues.38
or the vertical axis of the slide will be captured on the same We are also appreciative of and enthusiastic about methods that
spot. In imaging-based methods, the lack of cell boundaries validate cell types, not just to predict transcriptomic states, but
makes it hard to associate individual mRNA molecules with the more importantly to provide more possibilities for multi-omic
correct cells. In such methods, DAPI-stained nuclei may be the research. For example, some sequencing-based methods with
only method to determine cell outlines, especially when differ- high spatial resolution, such as Stereo-seq, involve the rapid
ent cell types are very close together. In Sci-Space, which lever- staining and imaging of just the tissue nuclei to avoid mRNA
ages sci-Plex to transfer DNA oligos into permeabilized nuclei, degradation.40 Simultaneously retaining the mRNA, protein or
DAPI-stained nuclei and spatial coordinates of oligo transfer are metabolites in the same cells may be very challenging. Proper
co-registered. The nuclei from the tissue on the slide are also tissue preparation and fixation are essential to preserve the tis-
extracted to enable sci-RNA-seq.29 Although Sci-Space retains sue, and so existing workflows containing these steps will need
a single-cell resolution, it is hard to avoid contamination from to be optimized to suit multimodal research.
adjacent cells. Moreover, the nuclei-assembling strategy may
collapse when applied to tissues that contain irregularly shaped
cells, such as neurons and glial cells in the brain. Regardless, ACKNOWLEDGMENTS
we expect that more robust approaches will ultimately permit This work was supported by grants from the Ministry of
the characterization of the whole transcriptome of single cells. Science and Technology of China (2021YFA1100900 and
The elaboration of some cytomembrane-outlining strategies, 2020YFE0203000), the National Natural Science Foundation of
such as immunofluorescent staining or auto-fluorescent reporter China (81730006, 81922002, 81861148029, and 81870086),
animals to visualize the plasma membrane, may help define cell CAMS Innovation Fund for Medical Sciences (2021-I2M-1-040
boundaries and realize the potential of single-cell SRT. Existing and 2021-I2M-1-019), Haihe Laboratory of Cell Ecosystem
www.blood-science.org  11
Duan et al

Innovation Fund (HH22KYZX0016) and Distinguished Young [30] Espina V, Wulfkuhle JD, Calvert VS, et al. Laser-capture microdissec-
Scholars of Tianjin (19JCJQJC63400). tion. Nat Protoc 2006;1:586–603.
[31] Nichterwitz S, Benitez JA, Hoogstraaten R, Deng Q, Hedlund E. LCM-
Seq: a method for spatial transcriptomic profiling using laser capture
microdissection coupled with PolyA-Based RNA sequencing. Methods
REFERENCES Mol Biol 2018;1649:95–110.
[1] Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome [32] Reuter JA, Spacek DV, Pai RK, Snyder MP. Simul-seq: combined DNA
Biol 2017;18:83. and RNA sequencing for whole-genome and transcriptome profiling.
[2] Tang F, Barbacioru C, Wang Y, et al. mRNA-Seq whole-transcriptome Nat Methods 2016;13:953–958.
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

analysis of a single cell. Nat Methods 2009;6:377–382. [33] Chen J, Suo S, Tam PP, Han JJ, Peng G, Jing N. Spatial transcriptomic
[3] van Galen P, Hovestadt V, Wadsworth Ii MH, et al. Single-cell RNA-Seq analysis of cryosectioned tissue samples with Geo-seq. Nat Protoc
reveals AML hierarchies relevant to disease progression and immunity. 2017;12:566–580.
Cell 2019;176:1265–1281.e24. [34] Picelli S, Faridani OR, Björklund AK, Winberg G, Sagasser S, Sandberg
[4] Han X, Zhou Z, Fei L, et al. Construction of a human cell landscape at R. Full-length RNA-seq from single cells using Smart-seq2. Nat Protoc
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

single-cell level. Nature 2020;581:303–309. 2014;9:171–181.


[5] Zheng GX, Terry JM, Belgrader P, et al. Massively parallel digital tran- [35] Picelli S, Björklund K, Faridani OR, Sagasser S, Winberg G, Sandberg
scriptional profiling of single cells. Nat Commun 2017;8:14049. R. Smart-seq2 for sensitive full-length transcriptome profiling in single
[6] Macosko EZ, Basu A, Satija R, et al. Highly parallel genome-wide cells. Nat Methods 2013;10:1096–1098.
expression profiling of individual cells using nanoliter droplets. Cell [36] Medaglia C, Giladi A, Stoler-Barak L, et al. Spatial reconstruction of
2015;161:1202–1214. immune niches by combining photoactivatable reporters and scRNA-
[7] Moor AE, Itzkovitz S. Spatial transcriptomics: paving the way for tis- seq. Science 2017;358:1622–1626.
sue-level systems biology. Curr Opin Biotechnol 2017;46:126–133. [37] Salmen F, Ståhl PL, Mollbrink A, et al. Barcoded solid-phase RNA cap-
[8] Marx V, Method of the Year 2020: spatially resolved transcriptomics. ture for Spatial Transcriptomics profiling in mammalian tissue sections.
Nat Methods 2021;18:1. Nat Protoc 2018;13:2501–2534.
[9] Femino AM, Fay FS, Fogarty K, Singer RH. Visualization of single [38] Liu Y, Yang M, Deng Y, et al. High-spatial-resolution multi-omics
RNA transcripts in situ. Science 1998;280:585–590. sequencing via deterministic barcoding in tissue. Cell 2020;183:1665–
[10] Levsky JM, Singer RH. Fluorescence in situ hybridization: past, present 1681.e18.
and future. J Cell Sci 2003;116:2833–2838. [39] Vickovic S, Eraslan G, Salmén F, et al. High-definition spatial transcrip-
[11] Raj A, van den Bogaard P, Rifkin SA, van Oudenaarden A, Tyagi S. tomics for in situ tissue profiling. Nat Methods 2019;16:987–990.
Imaging individual mRNA molecules using multiple singly labeled [40] Chen A, Liao S, Cheng M, et al. Spatiotemporal transcriptomic atlas of
probes. Nat Methods 2008;5:877–879. mouse organogenesis using DNA nanoball patterned arrays. bioRxiv
[12] Buxbaum AR, Haimovich G, Singer RH. In the right place at the right 2021. doi: https://doi.org/10.1101/2021.01.17.427004
time: visualizing and understanding mRNA localization. Nat Rev Mol [41] Cho CS, Xi J, Si Y, et al. Microscopic examination of spatial transcrip-
Cell Biol 2015;16:95–109. tome using Seq-Scope. Cell 2021;184:3559–3572.e22.
[13] Kejiou NS, Palazzo AF. mRNA localization as a rheostat to regulate sub- [42] Larsson C, Koch J, Nygren A, et al. In situ genotyping individual DNA
cellular gene expression. Wiley Interdiscip Rev RNA 2017;8:10.1002/ molecules by target-primed rolling-circle amplification of padlock
wrna.1416. probes. Nat Methods 2004;1:227–232.
[14] Holt CE, Bullock SL. Subcellular mRNA localization in animal cells [43] Gyllborg D, Langseth CM, Qian X, et al. Hybridization-based in situ
and why it matters. Science 2009;326:1212–1216. sequencing (HybISS) for spatially resolved transcriptomics in human
[15] Dermit M, Dodel M, Lee FCY, et al. Subcellular mRNA localization reg- and mouse brain tissue. Nucleic Acids Res 2020;48:e112.
ulates ribosome biogenesis in migrating cells. Dev Cell 2020;55:298– [44] Lee JH, Daugharthy ER, Scheiman J, et al. Highly multiplexed subcel-
313.e10. lular RNA sequencing in situ. Science 2014;343:1360–1363.
[16] Lubeck E, Cai L. Single-cell systems biology by super-resolution imag- [45] Lee JH, Daugharthy ER, Scheiman J, et al. Fluorescent in situ sequenc-
ing and combinatorial labeling. Nat Methods 2012;9:743–748. ing (FISSEQ) of RNA for gene expression profiling in intact cells and
[17] Lubeck E, Coskun AF, Zhiyentayev T, Ahmad M, Cai L. Single- tissues. Nat Protoc 2015;10:442–458.
cell in situ RNA profiling by sequential hybridization. Nat Methods [46] Wang X, Allen WE, Wright MA, et al. Three-dimensional intact-tissue
2014;11:360–361. sequencing of single-cell transcriptional states. Science 2018;361:eaat5691.
[18] Shah S, Lubeck E, Zhou W, Cai L. In situ transcription profiling of sin- [47] Foley JW, Zhu C, Jolivet P, et al. Gene expression profiling of single
gle cells reveals spatial organization of cells in the mouse hippocampus. cells from archival tissue with laser-capture microdissection and Smart-
Neuron 2016;92:342–357. 3SEQ. Genome Res 2019;29:1816–1825.
[19] Chen KH, Boettiger AN, Moffitt JR, Wang S, Zhuang X. Spatially [48] Meylan M, Petitprez F, Becht E, et al. Tertiary lymphoid structures
resolved, highly multiplexed RNA profiling in single cells. Science generate and propagate anti-tumor antibody-producing plasma cells in
2015;348:aaa6090. renal cell cancer. Immunity 2022;55:527–541.e5.
[20] Moffitt JR, Zhuang X. RNA imaging with multiplexed error-robust fluores- [49] Zeng Z, Li Y, Li Y, Luo Y. Statistical and machine learning methods
cence in situ hybridization (MERFISH). Methods Enzymol 2016;572:1–49. for spatially resolved transcriptomics data analysis. Genome Biol
[21] Chen F, Tillberg PW, Boyden ES. Optical imaging. Expansion micros- 2022;23:83.
copy. Science 2015;347:543–548. [50] Edsgard D, Johnsson P, Sandberg R. Identification of spatial expres-
[22] Coskun AF, Cai L. Dense transcript profiling in single cells by image sion trends in single-cell gene expression data. Nat Methods
correlation decoding. Nat Methods 2016;13:657–660. 2018;15:339–342.
[23] Eng CL, Lawson M, Zhu Q, et al. Transcriptome-scale super-resolved [51] Svensson V, Teichmann SA, Stegle O. SpatialDE: identification of spa-
imaging in tissues by RNA seqFISH. Nature 2019;568:235–239. tially variable genes. Nat Methods 2018;15:343–346.
[24] Trapnell C. Defining cell types and states with single-cell genomics. [52] Sun S, Zhu J, Zhou X. Statistical analysis of spatial expression pat-
Genome Res 2015;25:1491–1498. terns for spatially resolved transcriptomic studies. Nat Methods
[25] Kruse F, Junker JP, van Oudenaarden A, Bakkers J. Tomo-seq: a 2020;17:193–200.
method to obtain genome-wide expression data with spatial resolution. [53] Zhu J, Sun S, Zhou X. SPARK-X: non-parametric modeling enables
Methods Cell Biol 2016;135:299–307. scalable and robust detection of spatial expression patterns for large
[26] Rodriques SG, Stickels RR, Goeva A, et al. Slide-seq: a scalable technol- spatial transcriptomic studies. Genome Biol 2021;22:184.
ogy for measuring genome-wide expression at high spatial resolution. [54] Anderson A, Lundeberg J. sepal: Identifying transcript profiles with spatial
Science 2019;363:1463–1467. patterns by diffusion-based modeling. Bioinformatics 2021;37:2644–2650.
[27] Holler K, Junker JP. RNA tomography for spatially resolved transcrip- [55] Hu J, Li X, Coleman K, et al. SpaGCN: integrating gene expression,
tomics (Tomo-Seq). Methods Mol Biol 2019;1920:129–141. spatial location and histology to identify spatial domains and spa-
[28] Stickels RR, Murray E, Kumar P, et al. Highly sensitive spatial tran- tially variable genes by graph convolutional network. Nat Methods
scriptomics at near-cellular resolution with Slide-seqV2. Nat Biotechnol 2021;18:1342–1351.
2021;39:313–319. [56] Moehlin J, Mollet B, Colombo BM, Mendoza-Parra MA. Inferring
[29] Srivatsan SR, McFaline-Figueroa JM, Ramani V, Saunders L. Massively biologically relevant molecular tissue substructures by agglomerative
multiplex chemical transcriptomics at single-cell resolution. Science clustering of digitized spatial transcriptomes with multilayer. Cell Syst
2020;367:45–51. 2021;12:694–705.e3.

12 www.blood-science.org
 Chin Assoc of Blood Sci

[57] Zhao E, Stone MR, Ren X, et al. Spatial transcriptomics at subspot [85] Wu R, Guo W, Qiu X, et al. Comprehensive analysis of spatial archi-
resolution with BayesSpace. Nat Biotechnol 2021;39:1375–1384. tecture in primary liver cancer. Sci Adv 2021;7:eabg3750.
[58] Yang Y, Shi X, Liu W, et al. SC-MEB: spatial clustering with hid- [86] Moncada R, Barkley D, Wagner F, et al. Integrating microarray-based
den Markov random field using empirical Bayes. Brief Bioinform spatial transcriptomics and single-cell RNA-seq reveals tissue archi-
2022;23:bbab466. tecture in pancreatic ductal adenocarcinomas. Nat Biotechnol
[59] Dong R, Yuan GC. SpatialDWLS: accurate deconvolution of spatial 2020;38:333–342.
transcriptomic data. Genome Biol 2021;22:145. [87] Ji AL, Rubin AJ, Thrane K, et al. Multimodal analysis of composi-
[60] Elosua-Bayes M, Nieto P, Mereu E, Gut I, Heyn H. SPOTlight: seeded tion and spatial architecture in human squamous cell carcinoma. Cell
NMF regression to deconvolute spatial transcriptomics spots with sin- 2020;182:497–514.e22.
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

gle-cell transcriptomes. Nucleic Acids Res 2021;49:e50. [88] Andersson A, Larsson L, Stenbeck L, et al. Spatial deconvolution of
[61] Cable DM, Murray E, Zou LS, et al. Robust decomposition of cell type HER2-positive breast tumors reveals novel intercellular relationships.
mixtures in spatial transcriptomics. Nat Biotechnol 2022;40:517–526. bioRxiv 2020. doi:10.1101/2020.07.14.200600.
[62] Biancalani T, Scalia G, Buffoni L, et al. Deep learning and alignment [89] Svedlund J, Strell C, Qian X, et al. Generation of in situ sequenc-
of spatially resolved single-cell transcriptomes with Tangram. Nat ing based OncoMaps to spatially resolve gene expression profiles of
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

Methods 2021;18:1352–1362. diagnostic and prognostic markers in breast cancer. EBioMedicine


[63] Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accu- 2019;48:212–223.
rate integration of single-cell data with Harmony. Nat Methods [90] He B, Bergenstråhle L, Stenbeck L, et al. Integrating spatial gene
2019;16:1289–1296. expression and breast tumour morphology via deep learning. Nat
[64] Welch JD, Kozareva V, Ferreira A, Vanderburg C, Martin C, Macosko Biomed Eng 2020;4:827–834.
EZ. Single-cell multi-omic integration compares and contrasts fea- [91] Zugazagoitia J, Gupta S, Liu Y, et al. Biomarkers associated with
tures of brain cell identity. Cell 2019;177:1873–1887.e17. beneficial PD-1 checkpoint blockade in non-small cell lung cancer
[65] Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial recon- (NSCLC) identified using high-plex digital spatial profiling. Clin
struction of single-cell gene expression data. Nat Biotechnol Cancer Res 2020;26:4360–4368.
2015;33:495–502. [92] Gouin KH, 3rd, Ing N, Plummer JT, et al. An N-Cadherin 2 express-
[66] Abdelaal T, Mourragui S, Mahfouz A, Reinders MJ. T. SpaGE: spatial ing epithelial cell subpopulation predicts response to surgery, che-
gene enhancement using scRNA-seq. Nucleic Acids Res 2020;48:e107. motherapy and immunotherapy in bladder cancer. Nat Commun
[67] Shengquan C, Boheng Z, Xiaoyang C, Xuegong Z, Rui J. stPlus: a 2021;12:4906.
reference-based method for the accurate enhancement of spatial tran- [93] Peng G, Cui G, Ke J, Jing N. Using single-cell and spatial tran-
scriptomics. Bioinformatics 2021;37:i299–i307. scriptomes to understand stem cell lineage specification during
[68] Maseda F, Cang Z, Nie Q. DEEPsc: a deep learning-based map con- early embryo development. Annu Rev Genomics Hum Genet
necting single-cell transcriptomics and spatial imaging data. Front 2020;21:163–181.
Genet 2021;12:636743. [94] Stapel LC, Lombardot B, Broaddus C, et al. Automated detection
[69] Cang Z, Nie Q. Inferring spatial and signaling relationships between and quantification of single RNAs at cellular resolution in zebrafish
cells from single cell transcriptomic data. Nat Commun 2020;11:2084. embryos. Development 2016;143:540–546.
[70] Nitzan M, Karaiskos N, Friedman N, Rajewsky N. Gene expression [95] VanHorn S, Morris SA. Next-generation lineage tracing and fate map-
cartography. Nature 2019;576:132–137. ping to interrogate development. Dev Cell 2021;56:7–21.
[71] Ren X, Zhong G, Zhang Q, Zhang L, Sun Y, Zhang Z. Reconstruction [96] Beccari L, Moris N, Girgin M, et al. Multi-axial self-organization
of cell spatial organization from single-cell RNA sequencing properties of mouse embryonic stem cells into gastruloids. Nature
data based on ligand-receptor mediated self-assembly. Cell Res 2018;562:272–276.
2020;30:763–778. [97] van den Brink SC, Baillie-Johnson P, Balayo T, et al. Symmetry break-
[72] Arnol D, Schapiro D, Bodenmiller B, Saez-Rodriguez J, Stegle O. ing, germ layer specification and axial organisation in aggregates of
Modeling cell-cell interactions from spatial molecular data with spa- mouse embryonic stem cells. Development 2014;141:4231–4242.
tial variance component analysis. Cell Rep 2019;29:202–211.e6. [98] Peng G, Suo S, Cui G, et al. Molecular architecture of lineage allo-
[73] Kleshchevnikov V, Shmatko A, Dann E, et al. Cell2location maps cation and tissue organization in early mouse embryo. Nature
fine-grained cell types in spatial transcriptomics. Nat Biotechnol 2019;572:528–532.
2022;40:661–671. [99] van den Brink SC, Alemany A, van Batenburg V, et al. Single-cell and
[74] Li B, Zhang W, Guo C, et al. Benchmarking spatial and single-cell spatial transcriptomics reveal somitogenesis in gastruloids. Nature
transcriptomics integration methods for transcript distribution pre- 2020;582:405–409.
diction and cell type deconvolution. Nat Methods 2022;19:662–670. [100] Moris N, Anlas K, van den Brink SC, et al. An in vitro model of early
[75] Zeng H, Sanes JR. Neuronal cell-type classification: challenges, oppor- anteroposterior organization during human development. Nature
tunities and the path forward. Nat Rev Neurosci 2017;18:530–546. 2020;582:410–415.
[76] Codeluppi S, Borm LE, Zeisel A, et al. Spatial organization of [101] Liu C, Li R, Li Y, et al. Spatiotemporal mapping of gene expression
the somatosensory cortex revealed by osmFISH. Nat Methods landscapes and developmental trajectories during zebrafish embryo-
2018;15:932–935. genesis. Dev Cell 2022;57:1284–1298.e5.
[77] Moffitt JR, Bambah-Mukku D, Eichhorn SW, et al. Molecular, spa- [102] Chen A, Liao S, Cheng M, et al. Spatiotemporal transcriptomic atlas
tial, and functional single-cell profiling of the hypothalamic preoptic of mouse organogenesis using DNA nanoball-patterned arrays. Cell
region. Science 2018;362:eaau5324. 2022;185:1777–1792.e21.
[78] Maniatis S, Äijö T, Vickovic S. Braine C. Spatiotemporal dynam- [103] Asp M, Giacomello S, Larsson L, et al. A spatiotemporal organ-wide
ics of molecular pathology in amyotrophic lateral sclerosis. Science gene expression and cell atlas of the developing human heart. Cell
2019;364:89–93. 2019;179:1647–1660.e19.
[79] Chen WT, Lu A, Craessaerts K, et al. Spatial transcriptomics and in [104] Fawkner-Corbett D, Antanaviciute A, Parikh K et al. Spatiotemporal
situ sequencing to study Alzheimer’s disease. Cell 2020;182:976–991. analysis of human intestinal development at single-cell resolution. Cell
e19. 2021;184:810–826.e23.
[80] Janiszewska M. The microcosmos of intratumor heterogeneity: the [105] Hou X, Yang Y, Li P, et al. Integrating spatial transcriptomics and
space-time of cancer evolution. Oncogene 2020;39:2031–2039. single-cell RNA-seq reveals the gene expression profling of the human
[81] Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity embryonic liver. Front Cell Dev Biol 2021;9:652408.
and branched evolution revealed by multiregion sequencing. N Engl J [106] Crane GM, Jeffery E, Morrison SJ. Adult haematopoietic stem cell
Med 2012;366:883–892. niches. Nat Rev Immunol 2017;17:573–590.
[82] Hinohara K, Polyak K. Intratumoral heterogeneity: more than just [107] Golub R, Cumano A. Embryonic hematopoiesis. Blood Cells Mol Dis
mutations. Trends Cell Biol 2019;29:569–579. 2013;51:226–231.
[83] Berglund E, Maaskola J, Schultz N, et al. Spatial maps of prostate can- [108] Sturgeon CM, Ditadi A, Awong G, Kennedy M, Keller G. Wnt
cer transcriptomes reveal an unexplored landscape of heterogeneity. signaling controls the specification of definitive and primitive
Nat Commun 2018;9:2419. hematopoiesis from human pluripotent stem cells. Nat Biotechnol
[84] Thrane K, Eriksson H, Maaskola J, Hansson J, Lundeberg J. 2014;32:554–561.
Spatially resolved transcriptomics enables dissection of genetic het- [109] Xue Y, Liu D, Cui G, et al. A 3D atlas of hematopoietic stem and pro-
erogeneity in stage III cutaneous malignant melanoma. Cancer Res genitor cell expansion by multi-dimensional RNA-Seq analysis. Cell
2018;78:5970–5979. Rep 2019;27:1567–1578.e5.

www.blood-science.org  13
Duan et al

[110] Crosse EI, Gordon-Keylock S, Rybtsov S, et al. Multi-layered spa- [119] Shi L, Liu Y-H, Li K, et al. An AIE-based probe for rapid and ultrasen-
tial transcriptomics identify secretory factors promoting human sitive imaging of plasma membranes in biosystems. Angew Chem Int
hematopoietic stem cell development. Cell Stem Cell 2020;27:822– Ed Engl 2020;59:9962–9966.
839.e8. [120] Wang KC, Chang HY. Epigenomics: technologies and applications.
[111] Gao S, Shi Q, Zhang Y, et al. Identification of HSC/MPP expansion Circ Res 2018;122:1191–1199.
units in fetal liver by single-cell spatiotemporal transcriptomics. Cell [121] Deng Y, Bartosovic M, Kukanja P, et al. Spatial-CUT&Tag: spatially
Res 2022;32:38–53. resolved chromatin modification profiling at the cellular level. Science
[112] Calvanese V, Capellera-Garcia S, Ma F, et al. Mapping human haema- 2022;375:681–686.
topoietic stem cells from haemogenic endothelium to birth. Nature [122] Lee AC, Lee Y, Choi A, et al. Spatial epitranscriptomics reveals A-to-I
Downloaded from http://journals.lww.com/bls by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWn

2022;604:534–540. editome specific to cancer stem cell microniches. Nat Commun


[113] Zhang P, Li X, Pan C, et al. Single-cell RNA sequencing to track 2022;13:2540.
novel perspectives in HSC heterogeneity. Stem Cell Res Ther [123] Johnson CH, Ivanisevic J, Siuzdak G. Metabolomics: beyond
2022;13:39. biomarkers and towards mechanisms. Nat Rev Mol Cell Biol
[114] Tikhonova AN, Dolgalev I, Hu H, et al. The bone marrow microenvi- 2016;17:451–459.
YQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 01/08/2024

ronment at single-cell resolution. Nature 2019;569:222–228. [124] He J, Sun C, Li T, et al. A sensitive and wide coverage ambient mass
[115] Baryawno N, Przybylski D, Kowalczyk MS, et al. A cellular taxon- spectrometry imaging method for functional metabolites based molec-
omy of the bone marrow stroma in homeostasis and leukemia. Cell ular histology. Adv Sci (Weinh) 2018;5:1800250.
2019;177:1915–1932.e16. [125] Kompauer M, Heiles S, Spengler B. Atmospheric pressure MALDI
[116] Bigildeev AE, Petinati NA, Drize NJ. How methods of molecular biol- mass spectrometry imaging of tissues and cells at 1.4-mum lateral res-
ogy shape our understanding of the hematopoietic system. Mol Biol olution. Nat Methods 2017;14:90–96.
2019;53:626–637. [126] Yuan Z, Zhou Q, Cai L, et al. SEAM is a spatial single nuclear metab-
[117] Travlos GS. Normal structure, function, and histology of the bone olomics method for dissecting tissue microenvironment. Nat Methods
marrow. Toxicol Pathol 2006;34:548–565. 2021;18:1223–1232.
[118] Baccin C, Al-Sabah J, Velten L, et al. Combined single-cell and spatial [127] Sun C, Li T, Song X, et al. Spatially resolved metabolomics to dis-
transcriptomics reveal the molecular, cellular and spatial bone mar- cover tumor-associated metabolic alterations. Proc Natl Acad Sci USA
row niche organization. Nat Cell Biol 2020;22:38–48. 2019;116:52–57.

14 www.blood-science.org

You might also like