Single-cell sequencing to multi-omics technologies and applications
Single-cell sequencing to multi-omics technologies and applications
Abstract
Cells, as the fundamental units of life, contain multidimensional spatiotemporal information. Single-cell RNA
sequencing (scRNA-seq) is revolutionizing biomedical science by analyzing cellular state and intercellular
heterogeneity. Undoubtedly, single-cell transcriptomics has emerged as one of the most vibrant research fields
today. With the optimization and innovation of single-cell sequencing technologies, the intricate multidimensional
details concealed within cells are gradually unveiled. The combination of scRNA-seq and other multi-omics is at the
forefront of the single-cell field. This involves simultaneously measuring various omics data within individual cells,
expanding our understanding across a broader spectrum of dimensions. Single-cell multi-omics precisely captures
the multidimensional aspects of single-cell transcriptomes, immune repertoire, spatial information, temporal
information, epitopes, and other omics in diverse spatiotemporal contexts. In addition to depicting the cell atlas
of normal or diseased tissues, it also provides a cornerstone for studying cell differentiation and development
patterns, disease heterogeneity, drug resistance mechanisms, and treatment strategies. Herein, we review traditional
single-cell sequencing technologies and outline the latest advancements in single-cell multi-omics. We summarize
the current status and challenges of applying single-cell multi-omics technologies to biological research and
clinical applications. Finally, we discuss the limitations and challenges of single-cell multi-omics and potential
strategies to address them.
Keywords Single-cell multi-omics, scRNA-seq, scTCR-seq, scBCR-seq, Spatial transcriptomics, Proteomics,
Microbiome, Metabolome, Computational biology
Introduction
Transcriptomic analysis has provided fundamental
insights into the study of gene expression in exploring
the functions related to the process of life development,
†
disease progression, and drug action, etc. Over the past
Xiangyu Wu and Xin Yang contributed equally to this work.
decade, bulk RNA sequencing has shed light on biologi-
*Correspondence: cal functions from a pooled cell population transcrip-
Hongqian Guo
[email protected] tomic perspective. However, it represents an average
Rong Yang across the myriad of cells within a tissue, merely reflect-
[email protected] ing the characteristics of cell populations or perhaps pre-
1
Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital
of Medical School, Nanjing University, 321 Zhongshan Road, dominantly the information of the most numerous cells.
Nanjing 210008, Jiangsu, China Moreover, bulk RNA sequencing neither elucidates the
2
Medical School, Nanjing University, Nanjing, China variations of a sample at the single-cell level nor reflects
3
Department of Oncology, Nanjing Drum Tower Hospital, Affiliated
Hospital of Medical School, Nanjing University, Nanjing, China the expression levels of rare cells.
© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use,
sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included
in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The
Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available
in this article, unless otherwise stated in a credit line to the data.
Wu et al. Biomarker Research (2024) 12:110 Page 2 of 28
To tackle these constraints, scRNA-seq has revolution- single-cell analysis, providing technical support to unveil
ized biomedical science by single-cell expression pro- the secrets of cells.
filing. This technology enables detailed exploration of Single-cell multi-omics technologies have emerged.
genetic information at the cellular level across various They refer to the simultaneous measurement of various
tissues and diseases, capturing the inherent heterogeneity types of data in the same cell, allowing for an accurate
within samples. Since Tang et al. pioneered sequencing and detailed depiction of the cellular state. Integrating
technology on a single cell in 2009 [1], the methodology single-cell transcriptomic sequencing with comprehen-
has undergone continuous refinement and maturation. sive multi-omics data to map their inherent connections
These advancements have facilitated the development represents a critical and inevitable trend toward a more
of full-length transcriptome profiling, high-throughput nuanced, multidimensional understanding of life devel-
capabilities, and high-sensitivity scRNA-seq [2–4]. opment and the mechanisms underlying diseases.
Although well-established scRNA-seq has achieved These cutting-edge methods break through the limita-
great success and wide applications in the research field, tions of the conventional scRNA-seq, offering an excit-
it has also triggered new thinking because of its limita- ing solution to explore how cellular modalities affect cell
tions. Cellular information extends well beyond RNA state and function. Single-cell T cell receptor sequencing
sequencing, encompassing the genome, epigenome, pro- (scTCR-seq) [5] and single-cell B cell receptor sequenc-
teome, metabolome, etc., along with crucial details about ing (scBCR-seq) [6] effectively delineate the reper-
spatial relationships and dynamic alterations (Fig. 1B). toires of T and B cells, respectively, which can reveal
Therefore, scientists continually explore new methods for the immune system state. Integration with single-cell
proteomics (CITE-seq) enriches the information with
Fig. 1 (A) Conventional scRNA-seq technologies. (B) Overview of the single-cell multi-omics scheme
Wu et al. Biomarker Research (2024) 12:110 Page 3 of 28
proteomics that is both similarities and discrepancies Seurat, SingleCellExperiment, and SingleR, while
with the transcriptome [7]. Coupled with single-cell assay Python-based representative software includes Scanpy,
for transposase-accessible chromatin using sequencing Loom, and AnnData. Data preprocessing involves imple-
(scATAC-seq), researchers gain insights into chromatin menting data quality control, aligning sequences to ref-
accessibility, identifying active regulatory sequences and erence genomes, and generating expression matrices.
potential transcription factors (TFs). Besides, decipher- Subsequent analyses typically utilize formats like Seurat,
ing temporal and spatial information at the single-cell SingleCellExperiment, AnnData, or Loom. The general
level is fundamental for biological research. Although analysis workflow includes (1) filtering data based on
most temporal data is inferred via computational biology doublets, mitochondrial content, erythrocytes, etc., (2)
technology or scRNA-seq atlas created at multiple time selecting features as highly variable genes, and (3) dimen-
points, the experimental method to unveil newly syn- sion reduction, including principal component analysis
thesized RNA is another way [8]. Spatial transcriptomics (PCA), uniform manifold approximation and projection
technologies merge tissue sectioning with single-cell (UMAP), or t-distributed stochastic neighbor embedding
sequencing to compensate for the inability of scRNA-seq (t-SNE). The advanced analyses aim to answer the bio-
to characterize spatial locations. It is worth mentioning logical questions. Clustering and annotation of cell types
that computational biology is necessary for integrating answer what kinds of cell types are there. At the gene
and analyzing single-cell multi-omics data. The informa- level, differentially expressed genes (DEGs) and gene
tion content varies across different modalities. How to enrichment, which includes gene ontology (GO), kyoto
integrate and process these data is of utmost importance. encyclopedia of genes and genomes (KEGG), and gene
The standard workflow to analyze the multimodal data- set variation analysis (GSVA), aim to identify the differ-
sets is necessary for a broadly applicable strategy of sin- ential genes between cell types or specimens, as well as
gle-cell multi-omics. Additionally, computational biology to clarify their associated pathway profiles. Inferring TFs
methods are already widely employed to simulate related by single-cell regulatory network inference and cluster-
different dimension information. ing (SCENIC) provides the essential gene regulatory net-
The holistic view created by single-cell multi-omics work [12]. At the cell level, cell-cell communication offers
technologies is crucial for understanding the complexi- speculation from the perspective of cellular interactions
ties of biology, providing insights into cellular diversity, [13] while cell trajectory analysis incorporates the tem-
disease mechanisms, and potential therapeutic targets. poral information (Fig. 1A) [14]. Furthermore, inferring
In 2019, single-cell multimodal omics was selected as copy number variations (CNVs) with inferCNV analysis
Method of the Year [9]. In this review, we encapsulate the by comparing gene expression levels in cells with those in
advancements and applications of traditional scRNA-seq, a reference genome is particularly crucial in cancer biol-
outline various single-cell multi-omics methodologies, ogy [15].
and explore their biological and clinical implications, High cost and batch effects remain the major obstacles
while also contemplating current limitations and future for large cohort studies on scRNA-seq. To overcome
directions. these constraints, the integration of multiple samples
for large-scale scRNA-seq analysis has become a preva-
Conventional single-cell sequencing lent practice in research. Batch effects, hampering data
scRNA-seq, stepping onto the stage of history, has com- integration, may arise from different experimental con-
pletely reshaped the study approach of the complexity ditions, such as varying chips, sequencing lanes, or tim-
and heterogeneity within individual cells. scRNA-seq ing of cell processing. Integrating data from multiple
technologies mainly involve microfluidic chips, micro- experiments requires the use of employ algorithms such
droplets, and microwell-based approaches, which as Seurat’s canonical correlation analysis (CCA), mutual
have been well-introduced and compared in previ- nearest neighbors (MNN), or Harmony to batch correc-
ous articles [10, 11]. The main experimental steps of tion [16, 17].
scRNA-seq encompass preparing single-cell suspension, Sample multiplexed scRNA-seq is another solution,
isolating individual cells, capturing their mRNA, con- establishing an efficient method for massively paral-
ducting reverse transcription and nucleic acid amplifica- lel species-mixing experiments [18–22]. Currently, the
tion, and building a transcriptome library (Fig. 1A). predominant approach for this technology involves tag-
Analysis of scRNA-seq via bioinformatics is another ging individual samples with DNA oligonucleotides (oli-
cornerstone for visualizing and understanding the under- gos) barcodes before pooling them together, including
lying patterns and insights within the data. Tools for lipid-tagged DNA [21], chemical cross-linking reaction
analyzing scRNA-seq data are written in a variety of pro- [19], and genetic barcodes [20]. This technology is prag-
gramming languages, with R and Python being the most matic, multiplexing via DNA oligos and demultiplexing
prominent. R-based representative software includes conducted via bioinformatics independently of genetic
Wu et al. Biomarker Research (2024) 12:110 Page 4 of 28
background and sample origin, and is compatible with New advances in single-cell sequencing
other omics technologies, ensuring that the unique bio- Although high-throughput single-cell sequencing has
logical characteristics of individual samples are main- revealed the gene expression characteristics within the
tained. Consequently, it inherently avoids batch effects majority of physiological and diseased cells, the com-
or the loss of biological characteristics post-debatching. plexity of cells extends far beyond its scope. There is
Recently, Zhao et al. reported an improved ClickTags still a need to understand and decode the secrets of cells
method to enable the use in live-cell samples empowered from multiple dimensions. With the thriving develop-
by “click chemistry”, and eliminated the requirement for ment of biology, chemistry, bioinformatics, and other
methanol fixation of samples (Fig. 2B). Moreover, this advanced technologies, researchers are continuously
method has been successfully utilized across various developing new technologies and methods for single-cell
murine cells and human samples of bladder cancer that sequencing. Here, we summarize new advances in sin-
have undergone freeze-thaw cycles, demonstrating its gle-cell sequencing, which can be major trends in future
applicability to diverse single-cell specimens [22]. development.
Fig. 2 A brief method and principle of single-cell multi-omics technologies for (A) scTCR/BCR-seq, (B) ClickTag, (C) proteome, (D) microbiome, (E) me-
tabolome, (F) epigenome
Wu et al. Biomarker Research (2024) 12:110 Page 5 of 28
Fig. 3 The principles of key technologies in the dimensions of (A) time and (B) space at the single-cell level
Wu et al. Biomarker Research (2024) 12:110 Page 6 of 28
Palantir is a pseudo-time algorithm based on stochas- core feature of transcription dynamics at the single-cell
tic processes that, through dimensionality reduction and level. To address the constraint prohibiting large-scale
manifold analysis, effectively captures the continuity of scSLAM-seq, researchers developed new approaches that
cell states and models the transition from low-differen- integrate high-throughput unique molecular identifier
tiation cells to terminally differentiated cells [27]. It can (UMI)-based scRNA-seq analysis with metabolic label-
be applied to a variety of tissue types and is particularly ing. It has successfully enabled the acquisition of new/
well-suited for addressing less-studied differentiation old transcriptomes from thousands of single cells. It has
systems. been applied in transcription factor activity and cell state
CytoTRACE leverages gene counting and expression trajectories during neuronal activation [33], as well as
to reconstruct cell trajectories, enabling the prediction of the transcriptional dynamics in colorectal cancer cells
relative differentiation states of single cells based on sin- treated with DNA-demethylating drugs [34]. We antici-
gle-cell RNA expression data, without being constrained pate the potential of dynamic transcriptomics application
by specific time scales or the presence of continuous in response to external stimuli (including viral infections
developmental processes in the data [28]. Additionally, and pharmacological interventions), embryonic devel-
CytoTRACE is independent of tissue type, species, and opment, cell differentiation, tumor progression, and
platform and can be used to predict differentiation status immune cell transformation is immense. So far, the appli-
in scRNA-seq data without any prior information. cation of these technologies in vivo remains unexplored,
despite their established utility at the bulk level [35]. It
Experimental method should be emphasized that the combination of scRNA-
As mentioned above, trajectory inference for single- seq with metabolic labeling in vivo is a feasible avenue
cell analysis provides an avenue to explore the temporal that warrants further identification.
information about cells. The fundamental limitation is In addition to 4sU, Battich et al. presented an approach
that it is only a computational inference and cannot rep- that utilizes 5-ethynyl-uridine (EU) and click chemistry
resent the actual existence of dynamic RNA processes. to separate new and old RNA, thus providing a dynamic
Emerging experimental technologies allow us to distin- view of RNA synthesis and turnover [36]. 5-Ethynyl-
guish time-resolved phenomena in reality by chemical or 2-deoxyuridine (EdU), a thymidine analog, was applied
biological methods (Table 1). for scRNA-seq and scATAC-seq dynamics [37]. Meta-
bolic labeling-based RNA velocity can accurately reca-
Metabolic RNA labeling pitulate the dynamics of gene expression.
Metabolic RNA labeling effectively integrates with high-
throughput scRNA-seq, addressing limitations pertaining Fluorescent timer
to RNA transcription dynamics [8]. The technology was The research by Gehart et al. showcased that using fluo-
initially applied at the bulk RNA level [30]. Currently, the rescent reporters offers a promising way to reveal real-
most typical metabolic RNA labeling in scRNA-seq is a time-resolved scRNA-seq [38]. Neurog3 is transiently
nucleoside analog called 4-thiouridine (4sU) [31, 32]. The expressed in enteroendocrine (EE) progenitor cells.
brief technical principle is as follows: After tissue dissoci- They engineered the integration of three independent
ation, the culture medium is supplementary with 4sU. In proteins: NEUROG3, dTomato (red), and destabilized
the process of nascent RNA synthesis, 4sU replaces uracil mNeonGreen (green) to study the dynamics of EE cell
(U). Reverse transcriptase misread 4sU as cytosine (C). development. The differing decay rates of mNeonGreen
This misreading leads to incorrect pairing, where adenine and dTomato enable the measurement of the actual time
(A), which normally pairs with U, is replaced by guanine elapsed since Neurog3 expression in individual cells,
(G). This ultimately results in T-to-C substitutions in the determined by the red: green fluorescence ratio. Integrat-
labeled new RNA. ing this approach with scRNA-seq, they have successfully
Single-cell, thiol-(SH)-linked alkylation of RNA for created a real-time-resolved map, revealing the intri-
metabolic labelling sequencing (scSLAM-seq) pioneered cate process of EE cell differentiation and development.
the application of the metabolic labeling method at the Recently, Kirschenbaum et al. developed fluorescent-
single-cell level [32]. This breakthrough identified DEGs based dynamics scRNA-seq called Zmen-seq in vivo,
in mouse fibroblasts infected and uninfected with cyto- which uncovered immune dysfunctional trajectories of
megalovirus (CMV) by distinguishing between total, old, glioblastoma [39].
and new RNA matrices. The study highlighted a critical
insight: most DEGs are primarily detectable in new RNA, CRISPR/Cas9
eluding detection in analyses of old or total RNA. Addi- Lineage tracing can be deduced by the patterns of muta-
tionally, it revealed that interferon and NF-κB varied sig- tions shared between cells. The emergence of CRISPR/
nificantly with different levels of infection, which was a Cas9 technology facilitates the deliberate introduction
Table 1 Time-resolved single-cell technologies based on experimental method
Experimental Pub- Principle Advantages and disadvantages Cultivation Research object Number of Ref-
Method lished method cells er-
Wu et al. Biomarker Research
date ence
scSLAM-seq 2019 Jul 4sU Advantages: captures transient and dynamic nature of transcription; applicable for studying single- In vitro lytic mouse 110 [32]
cell response to perturbations cytomegalovirus
Disadvantages: single cell sample size is too small; extracting mRNA from cells to perform chemical infection
conversion
NASC-seq 2019 Jul 4sU Advantages: identifying newly synthesized RNA In vitro K562 and Jurkat < 200 [44]
(2024) 12:110
Disadvantages: single cell sample size is too small; extracting mRNA from cells to perform chemical cells
conversion; restricted to cell populations
Sci-fate 2020 4sU Advantages: using combinatorial indexing to detect more cells; a greater number of genes detected In vitro Cell cycle and > 6,000 [45]
Aug per cell; in situ 4sU (high reaction efficiency and low mRNA loss) glucocorticoid
Disadvantages: requires complex experimental setups, leading to cell loss receptor activation
scNT-seq 2020 4sU Advantages: droplet microfluidics, high-throughput and UMI-based In vitro Neuronal activation ∼ 55,000 [33]
Oct Disadvantages: limited sequencing coverage for scRNA-seq; cannot wash away cell-free RNAs in the
droplets
Well-TEMP-seq 2023 4sU Advantages: low cell loss rate, high-throughput and cost-effective; In vitro Anticancer drugs 16,000 [34]
Mar Disadvantages: only for in vitro experiment treat colorectal
cancer cells
scEU-seq 2020 EU Advantages: high UMI; high-throughput In vitro Intestinal organoids 5,422 [36]
Mar Disadvantages: requires specialized reagents and procedures; physical separation of new and old
transcripts to conduct two libraries
TrackerSci 2023 EdU Advantages: identifying newborn brain cells; revealing cell dynamics In vitro/In brain newborn cells ∼ 800,000 [37]
Seq Disadvantages: a lesser detection of the quiescent cell population vivo
Fluorescent timer 2019 dTomato (red); Advantages: offers real-time tracking of gene expression changes during the biological process; In vitro/in Mice and organoids / [38]
Feb destabilized from instantaneous changes to long-term process vivo
mNeonGreen
(green)
Zmen-seq 2024 fluorophore Advantages: exploring biological processes in vivo for multiple days; In vivo Immune cells in 8,976 [39]
Jan pulse labels Disadvantages: the number of fluorescent timers is limited tumor environment
(TME)
CARLIN 2020 CRISPR-Cas9 Advantages: Combines CRISPR-Cas9 with scRNA-seq for lineage tracing In vivo Genetic lineage / [41]
Jun tracing
Page 7 of 28
Wu et al. Biomarker Research (2024) 12:110 Page 8 of 28
of mutations using guide RNAs (gRNAs) [40]. These technologies into three classes (Table 2) and present the
induced mutations can be applied to readout of lin- primary technologies and principles (Fig. 3B).
eage histories at the single-cell level [41]. This advanced
approach is used to trace cell lineage dynamics during Image-based in situ technologies
embryonic development and reconstruction [42], as well The image-based in situ technologies evolve from in
as to track the growth and dissemination of lung tumor situ hybridization (ISH) and in situ sequencing (ISS).
xenografts in mice [43]. ISH employs labeled probes to hybridize with target
sequences in cells, thereby visualizing the locations of
Spatial information the sequences [49]. ISH-based technologies use com-
Despite significant advancements in scRNA-seq, current plementary fluorescent probes to hybridize with target
scRNA-seq technologies require isolating cells from the sequences and detect them. Single-molecule fluorescence
tissues, resulting in the loss of spatial information, which ISH (smFISH) is a high-resolution technology that uti-
is crucial for investigating intercellular interactions and lizes multiple short oligonucleotide probes coupled with
functional relevance [46, 47]. In recent years, thriving fluorescent moieties to selectively detect transcripts
spatial omics technologies have provided robust tools [50], but it is limited by the throughput of detection.
for analyzing spatial information of cells, with spatially The emerging multiplexed FISH technologies address
resolved transcriptomics acknowledged as Method of the limitation. Sequential FISH (seqFISH) barcodes the
the Year in 2020 [48]. Here, we categorize spatial omics transcripts fixed within cells through multiple rounds of
hybridization, imaging, and probe removal, allowing for
the detection of the entire transcriptome through four sections for subsequent analysis. Laser capture micro-
dyes and eight rounds of hybridization [51]. Nevertheless, dissection (LCM) is a typical technology capable of cap-
it suffers optical crowding when profiling an excessive turing ROIs at resolutions ranging from cell-population
number of transcripts [52]. seq-FISH + is an improved to single-cell via laser cutting [65]. Geographical posi-
seqFISH, profiling 10,000 genes within individual cells by tion sequencing (Geo-seq), an integration of LCM and
expanding the barcode base palette to 60 pseudo colors scRNA-seq, enables the construction of 3D transcrip-
[53]. In addition, it only labels a fraction of transcripts tome atlases, thereby revealing cellular heterogeneity and
during each hybridization cycle to avoid optical crowd- spatial disparities [66]. Additionally, the integration of
ing. In another strategy, multiplexed error-robust FISH high-content imaging, LCM, and multiplexed mass spec-
(MERFISH) can simultaneously image 100 to 1,000 kinds trometry can extend single-cell proteomics to intact tis-
of RNA within individual cells by assigning combinatorial sue, significantly improving biological insight [67]. These
FISH labeling to RNA, followed by successive hybridiza- studies suggest that LCM is a promising technology for
tion and imaging [54]. isolating ROIs within tissues for multi-omics analysis.
ISS, first reported in 2013, utilized padlock probes to GeoMx digital spatial profiler (DSP), a commercially
bind with cDNA produced by reverse transcription, available technology, is capable of spatially profiling RNA
followed by rolling-circle amplification (RCA) to pro- or proteins within the ROIs [68]. It uses a photocleavable
duce rolling-circle products (RCP), which were in situ (PC) linker to connect oligo sequence (DSP barcodes)
sequenced and imaged within tissues or cells [55]. Cur- with RNA probe or antibody in fixed tissue, selecting
rently, fluorescent in situ sequencing (FISSEQ) and ROIs, and releasing barcodes with UV for sequencing.
spatially-resolved transcript amplicon readout mapping This technology has been used to dissect the heterogene-
(STARmap) are two representatives of ISS-based tech- ity of glomerular transcriptional profiler missed by LCM
nologies. FISSEQ is an untargeted strategy that uses in collapsing glomerulopathy [69], as well as to identify
hexamer primers to reverse transcribe RNA in fixed biomarkers associated with immune checkpoint inhibitor
cells, followed by cDNA circularization, and generates (ICI) resistance in non-small cell lung cancer (NSCLC)
a sequencing library, further improving the detection [70]. However, these technologies are limited by through-
throughput of ISS [56]. STARmap hybridizes directly to put, as each ROI requires individual collection and
mRNA with a SNAIL probe and then undergoes RCA to processing.
obtain DNA amplicons, thereby avoiding reverse tran-
scription [57]. The distinctive feature is its capability to Spatial barcode-based technologies
achieve in situ sequencing in three-dimensional (3D) The core of spatial barcode-based technologies is cap-
intact tissue. STARmap PLUS further improves detection turing transcripts within tissues or cells using spatial
throughput and is compatible with both transcriptome barcodes (DNA oligos) array on glass slides [71]. Sub-
sequencing and protein detection within the same tissue sequently, library preparation and next-generation
section, providing a more comprehensive insight into the sequencing (NGS) are performed.
biological systems [58]. Spatial transcriptomics (ST) was a milestone achieve-
The image-based in situ technologies can achieve sub- ment, carrying epochal significance for spatially resolved
cellular resolution [59]. When combined with single-cell transcriptomics [72]. It innovatively captured polyad-
omics technologies, they hold great potential for diverse enylated RNA on spot-equipped slides, with each spot
applications, particularly in neuroscience. For instance, containing unique spatial barcodes, ensuring that each
Shi et al. employed STARmap PLUS on 20 central ner- transcript was precisely mapped back to its respec-
vous system (CNS) tissue sections and integrated the tive spot through the spatial barcode [73]. ST was first
resulting data with the published scRNA-seq atlas, applied in adult mouse olfactory bulb and human breast
achieving molecular cell typing of the CNS [60]. Yao et cancer, achieving RNA sequencing while preserving two-
al. integrated MERFISH and scRNA-seq to construct a dimensional spatial information. 10× Genomic improved
transcriptomic and spatial atlas of the mouse whole brain it and released 10× Visium, which possessed a higher
and built a platform to visualize these data, providing spatial resolution [74]. It has been commercialized and
precious resources for deciphering the complexity of the extensively employed across diverse fields such as devel-
mammalian brain [61]. Additionally, the combinations opmental biology [75], cancer biology [76], as well as
can be applied in hematology [62], stem cell research neuroscience [77, 78]. For instance, Olaniru et al. applied
[63], and developmental biology [64]. the integration of scRNA-seq with 10× Visium to the
developing human fetal pancreases, analyzing the differ-
ROI selection-based technologies entiation and maturation processes of various cell types
These technologies employ specific methodologies to [75]. Galeano Niño et al. applied 10× Visium to oral squa-
precisely select regions of interest (ROIs) from tissue mous cell carcinoma and colorectal cancer, identifying
Wu et al. Biomarker Research (2024) 12:110 Page 10 of 28
the identity and in situ position of the microbial commu- co-mapping transcriptome and epitope. A very recently
nities within the tumors [76]. Hasel et al. integrated bulk developed technology, Slide-tags, can label single nuclei
RNA-seq, and scRNA-seq with 10× Visium, uncovering with spatial barcodes and isolate them for multi-omics
spatiotemporal heterogeneity in the response of different analysis [88]. Altogether, these spatial multi-omics tech-
astrocyte subsets to inflammation in the brain [78]. These nologies provide more comprehensive biological infor-
applications suggest that 10× Visium is a highly promis- mation, deepening our understanding of the intricate
ing technology for dissecting spatial information. spatial and molecular interactions in the field of life sci-
Slide-seq captures transcripts on a slide equipped ence and biomedical research.
with random 10-µm DNA-barcoded beads and gains the
positions of barcodes via in situ indexing, enabling spa- Genome
tial transcriptomic analysis at a near-cellular resolution Although genomes are generally considered to be sta-
[79]. Stickels et al. optimized Slide-seq and reported the ble, there is still a small possibility of genetic mutations
highly sensitive Slide-seqV2, leading to a tenfold increase occurring with each DNA replication. Single-cell whole-
in RNA capture efficiency [80]. High-definition spatial genome sequencing (scWGS) is capable of elucidating
transcriptomics (HDST) utilizes a split-pool approach genomic heterogeneity and can therefore be utilized to
to generate a dense and barcoded bead array, capturing analyze genomic mutations in single cells. This technol-
RNA at a 2-µm resolution [81]. Recently developed spa- ogy involves the isolation of individual cells or nuclei fol-
tial enhanced resolution omics-sequencing (Stereo-seq) lowed by whole-genome amplification (WGA), library
utilized a randomly barcoded DNA nanoball patterned preparation, and sequencing [89]. It has been applied to
array chip to achieve single-cell resolution and has been uncover somatic mutations in multiple cell types, such
employed to delineate the spatiotemporal transcriptomic as human bronchial epithelial cells [90], and B lympho-
landscape of mouse organ development [82]. cytes [91], offering novel insights into the pathogenesis of
However, the aforementioned spatial barcode-based diseases.
technologies suffer several constraints, such as multi- Single nucleotide polymorphism (SNP), a common
cellular resolution, low sensitivity, and the necessity for form of genetic variation, is caused by the transition,
sequencing to obtain positional indexing. To address transversion, insertion, or deletion of individual bases.
these constraints, Cao et al. presented a dendrimeric Expression quantitative trait loci (eQTL) analysis can
DNA coordinate barcoding design for spatial RNA profile genetic variations (especially SNP) that affect
sequencing (Decoder-seq) [83]. The central innovation gene expression, offering enhanced insights into the
was to employ a microfluidics-assisted combinational relationship between genetic variants and gene regula-
barcoding approach to create high-density spatial bar- tion [92, 93]. The integration of scRNA-seq with eQTL
code arrays on a 3D dendrimeric nanosubstrate, enabling was first reported in 2018 [94]. Kang et al. utilized mul-
cost-effective, highly sensitive, near-cellular resolution tiplexed droplet scRNA-seq to profile eight immune cell
spatial transcriptomics research. They utilized it to spa- populations from 23 donors and subsequently conducted
tially resolve the distribution of low-expressed olfactory eQTL analysis, identifying 32 cis-eQTLs, 22 of which
receptor genes and accurately depict the spatial single- were cell-specific. Recently, Ding et al. constructed the
cell landscape of the hippocampus. first integrated human sceQTL database, which com-
Recently, spatially resolved multi-omics has been rec- prises ∼ 16 million SNPs and ∼ 0.69 million sceQTLs [95],
ognized as one of the noteworthy technologies in 2023 providing a valuable resource for disease susceptibility
[84]. Fan’s group pioneered spatial multi-omics technol- gene discovery.
ogy. They innovatively employed two orthogonal chips In addition, multiple multi-omics technologies for inte-
equipped with parallel microfluidic channels to deliver grative analysis of genome and transcriptome have been
DNA barcodes to tissue sections, developing the first developed, among which genome and transcriptome
spatial multi-omics technology, deterministic barcod- sequencing (G&T-seq) [96] and gDNA-mRNA sequenc-
ing in tissue for spatial omics sequencing (DBiT-seq), ing (DR-seq) [97] stand out as two representative tech-
enabling simultaneous detection of the whole tran- nologies. G&T-seq utilizes oligo-dT-coated beads to
scriptome and 22 proteins [85]. Furthermore, the group isolate mRNA and DNA, which are subsequently ampli-
extended this approach to spatial ATAC-RNA-seq and fied and subjected to whole transcriptome sequencing
spatial assay of cleavage under targets and tagmentation and WGS, respectively. Whereas DR-seq employs a pre-
and RNA sequencing (spatial CUT&Tag-RNA-seq) [86], amplification-and-separation strategy to decouple DNA
which achieve co-profile of epigenome and transcrip- and mRNA molecular analytes, thereby avoiding physical
tome, as well as spatial co-indexing of transcriptomes nucleic acid separation before amplification. Therefore,
and epitopes for multi-omics mapping by highly parallel compared with G&T-seq, DR-seq has a lower cross-con-
sequencing (spatial CITE-seq) [87], which is capable of tamination rate and a higher recovery rate. Collectively,
Wu et al. Biomarker Research (2024) 12:110 Page 11 of 28
these technologies hold vast potential for deciphering methods of scATAC-seq were developed in 2015, which
genetic variations and their impacts on gene expression. enabled the exploration of chromatin accessibility at the
single-cell level. The first one utilized a programmable
Epigenome microfluidics platform to capture single cells, followed
Epigenomics aims to explore how chemical modifica- by Tn5 transposase tagmentation and library amplifica-
tions and spatial structure alterations of the genome tion with cell-identifying barcoded primes [111]. The
affect gene function and expression regulation. Decipher- other one utilized an integrative method combining com-
ing the epigenomic features such as DNA methylation, binatorial cell indexing with ATAC-seq to analyze chro-
chromatin accessibility and histone modifications at the matin accessibility within over 15,000 cells [112]. 10×
single-cell level allows us to study cell lineages and differ- Genomics developed a Chromium platform and applied
entiation states [98]. it to scATAC-seq, which combined Tn5 transposase tag-
In eukaryotes, the most common DNA methylation mentation within bulk nuclei and single-nuclei isolation
occurs on the fifth carbon atom of C within the CpG through the droplet system [113]. Currently, the integra-
island, yielding 5-methylcytosine (5-mC). Bisulfite con- tion of scRNA-seq and scATAC-seq has been applied to
version is the gold standard for DNA methylation anal- explore the regulation of human developmental hemato-
ysis [99]. The primary principle is that methylated C in poiesis [114], as well as to find potential therapeutic tar-
DNA remains unchanged after treatment with bisulfite, gets for clear cell renal cell carcinoma [115].
whereas unmethylated C is converted to U [100]. After Histone modifications are chemical modifications that
PCR amplification and high-throughput sequencing, the occur at specific sites on histone molecules, thereby
bases that methylated can be ascertained by compari- affecting chromatin structure stability and gene expres-
son with reference sequences. Single-cell DNA methyla- sion regulation [116]. Chromatin immunoprecipitation
tion sequencing based on bisulfite conversion and NGS sequencing (ChIP-seq) is a common method to pro-
can be categorized into single-cell reduced representa- file histone modifications [117]. Single-cell ChIP-seq
tion bisulfite sequencing (scRRBS) [101] and single-cell (scChIP-seq) tags nucleosomes with barcodes via a drop-
whole-genome bisulfite sequencing (scWGBS) [102], let microfluidic platform before conventional ChIP-seq,
which achieve single-base resolution. These technologies and it was employed to interrogate the chromatin land-
play crucial roles in investigating cell differentiation and scapes of breast cancer [118]. However, ChIP-seq has a
development. The first single-cell multi-omics technology high demand for experimental samples. To address the
achieving co-profile of the DNA methylome and tran- obstacle, Kaya-Okur et al. introduced CUT&Tag, which
scriptome is single-cell methylome and transcriptome utilizes Protein A-Tn5 transposase to cleave the DNA
sequencing (scM&T-seq) [103], which utilizes G&T-seq sequences bound by targeted protein and integrates
to separate and amplify genomic DNA and mRNA from sequencing adapters with the cleaved sequences [119].
the same single cell and applies scBS-seq [104] to gener- On this basis, single-cell CUT&Tag (scCUT&Tag) inte-
ate DNA methylation data. In addition, single-cell triple grated CUT&Tag with 10× Genomics scATAC-seq pro-
omics sequencing (scTrio-seq), which co-profiles the tocol, enabling the investigation of histone modifications
genome, DNA methylome, and transcriptome through at single-cell resolution [120]. It was applied to explore
scRRBS and WGS, has been applied to 25 cancer cells the histone modification features of regulatory elements
and identified two distinct subsets [105]. However, one and gene bodies in the central nervous system cells of
constraint of these technologies is that bisulfite conver- mice. Recently, some multi-omics technologies have
sion involves intense chemical reactions that can lead been developed, such as Paired-Tag [121] and combined
to significant DNA degradation and consequent loss of assay of transcriptome and enriched chromatin binding
information. (CoTECH) [122], which utilize a combinatorial barcod-
Chromatin accessibility, a key epigenetic feature, plays ing strategy, achieving co-profile of histone modifications
a pivotal role in regulating gene expression by allowing and transcriptome in single cells.
transcriptional machinery to interact with regulatory ele-
ments, thereby facilitating the initiation or suppression Cellular protein and epitope
of gene transcription in open chromatin regions [106]. Single-cell proteomics is a more nascent field. Tran-
Recently a variety of technologies have been developed scriptomic features may not exhibit a comprehensive
to interrogate chromatin accessibility [107–109], whereas snapshot of cellular heterogeneity since similar gene
ATAC-seq barges to the forefront as a landmark break- expression profiles may be identifiable in other modali-
through that utilizes the Tn5 transposase to fragment ties that are simultaneously measured. Indeed, the tran-
open chromatin and labels the genome with sequencing scriptomes and proteomes represent distinct molecular
adaptors [110]. Subsequently, the labeled genome under- modalities, such as post-translational modifications that
goes PCR amplification and sequencing (Fig. 2F). Two cannot be captured by transcriptomics. It’s crucial to
Wu et al. Biomarker Research (2024) 12:110 Page 12 of 28
A series of heavyweight studies on the TILs have exhib- [145], conducting vaccine studies [146], etc., and proved
ited the T cell and TCR clonotype diversity, constructing invaluable in studies investigating the dynamics of the
the landscape of T cell heterogeneity and dynamics in the immune system. From a clinical perspective, it helps
TME [135–137]. diagnose and monitor tumor immunology [144], infec-
In routine analysis, the primary objective is to map tious diseases [145, 147], autoimmune diseases, and
and analyze the diversity of clonotypes or CDR3. Addi- immunodeficiencies.
tionally, clonotype overlap represents another critical
dimension, empowering the characterization of TCR Microbiome
sequences shared across different tissues or tracking of Microbiome refers to the collection of microorganisms
key clonotypes during therapeutic interventions. From living in a particular environment. The main technolo-
the excellent work of Zhang’s group, they pioneered the gies in the microbiome field are metagenomic sequenc-
development of multiple indicators such as single T cell ing and 16 S rRNA sequencing [148–150], updating our
analysis by RNA sequencing and TCR tracking (STAR- understanding of microbial community on a more micro-
TRAC) (for clonal expansion, tissue migration, and state scopic level (Fig. 2D). Recently an advanced technology
transition), Ro/e, and OR (for tissue distribution) to assess called barcoding bacteria for identification and quanti-
the status of TCR [135–137]. fication (BarBIQ), achieved precise single-base in 16 S
It is crucial to explore the differentiation of TCRs in cell rRNA sequencing through the use of unique barcodes
types/ tissues/ conditions. Chen et al. developed TCRdb, and a droplet-based approach [151]. Microbiomes can
a comprehensive TCR database, that aids in identify- be assessed at the single-cell level, addressing the issue
ing the states and functions of specific sequences [138]. where traditional methods may obscure the simultane-
Another significant challenge is unraveling the binding ous measurement of cell counts for each type of bacteria.
between TCR-pMHC in the context of infectious diseases In the context of scRNA-seq, although it has become a
or tumors. Advanced bioinformatic algorithms accelerate transformative technology for profiling gene expression
research breakthroughs in this field. Among these tools, levels in thousands of eukaryotic cells, challenges such as
NetMHCpan stands out in predicting the binding affin- the low volume of RNA, no polyadenylate tail in bacterial
ity between peptides and MHC-I/II, respectively [139]. RNA, and resistant cell wall have long hindered the adap-
Additionally, Gao et al. have developed a computational tation of scRNA-seq technology to microbes. To over-
tool to predict TCR-peptide binding via a machine learn- come these obstacles, Kuchina et al. introduced microbial
ing method [140]. split-pool ligation transcriptomics (microSPLiT), a tech-
In all, scTCR-seq and associated analyses enhance the nology that can identify the scarce subpopulations of
exploration and application of TCR sequencing. They cells down to a minuscule proportion of 0.142%, which
facilitate the exploration of mechanisms, helping cancer was crucial for revealing rare cellular states that are sig-
early-stage diagnosis, treatment selection, and progno- nificant from a physiological perspective [152]. Similarly,
sis prediction, and designing engineering therapeutic BacDrop was developed for bacterial scRNA-seq, identi-
strategies. fying bacterial types and quantifying the number of spe-
cific types of cells [153]. Besides, Microbe-seq applied
BCR microfluidic-droplet operation and bioinformatic analy-
Similar to TCR, BCR consists of immunoglobulin heavy sis to obtain the genomes of numerous microbes with
chains and light chains, the diversity of it is produced by single-cell resolution, and most single-amplified genomes
gene rearrangement of V(D)J gene. Based on its struc- had a purity of over 95% [154].
ture, BCR plays a central role in the adaptive immune The intratumor microbiome has emerged as a novel
response by recognizing specific antigens, which trig- and rapidly evolving research frontier, with the discovery
ger B cell activation, subsequently producing antibod- of microorganisms in various cancer types, including in
ies [141]. Around a decade ago, paired scBCR-seq was some organs traditionally considered to be sterile, pri-
introduced [142, 143]. scBCR-seq allows for a thorough marily in gastrointestinal cancers [155–157]. However,
analysis of BCR repertoires, providing crucial informa- the subtle relationship between them and cancer remains
tion about their diversity and function at the single-cell unclear, sequencing technologies may be an effective
level. More recently, Ian et al. challenged limited infor- solution. Galeano et al. introduced invasion-adhesion-
mation about BCR-seq to antigen specificity and devel- directed expression sequencing (INVADEseq) targeting
oped LInking B-cell Receptor to Antigen specificity a conserved area of the bacterial 16 S rRNA, enabling
through sequencing (LIBRA-seq) to confirm for HIV- the effective creation of cDNA libraries containing bac-
and influenza-specific antibodies [6]. Combined with terial transcripts derived from human cells associated
scRNA-seq, scBCR-seq was applied for producing cell with bacteria. The technology is crucial to uncovering the
atlas [144], identifying different neutralizing antibodies
Wu et al. Biomarker Research (2024) 12:110 Page 14 of 28
complexity of microbiota interactions within tumor tis- for complex systems [173]. This promising technology
sues [158, 159]. has been effectively integrated with RNA-seq and flow
Another interesting bioinformatical approach called cytometry. Its combination with sing-cell technology is
Single-cell Analysis of Host-Microbiome Interactions bound to reflect CCIs from a real perspective and could
(SAHMI) [160], is quite fascinating as it systematically offer real insight into the exploration of tumor-reactive
extracts real microbial signals and quantifies microbiome TCR.
profiles directly from mammalian host sequencing data.
SAHMI advances a similar method into the realm of sin- Perspective in application in biological research
gle-cell analysis, enabling the identification of microbial Currently, single-cell multi-omics technologies are rap-
species related to specific cell types and uncovering the idly evolving, offering robust tools for depicting intricate
relationship between microbial and transcriptome pro- cellular landscapes. They have been applied to a diversity
files, facilitating a deeper investigation into their contri- of fields including, but not limited to, cell atlas construc-
bution to intercellular communication networks [160]. tion, developmental biology, pathways identification, and
novel targets discovery, with remarkable achievements
Metabolome (Fig. 4).
Metabolome focuses on the biochemical reactions within
cells, encompassing the collection of all metabolites of Cell atlases construction
tissue in a specific physiological period and metabolic Constructing cell atlases is a common function of single-
features (Fig. 2E). The field of single-cell metabolo- cell multi-omics technologies. Cell atlases, as one of the
mics marks a pivotal era in unraveling the complexities seven noteworthy technologies in 2024 [175], can display
of cellular processes at the individual cell level. Mass detailed information about various cell types within dif-
spectrometry (MS) [161–164] and nuclear magnetic ferent organisms, allowing for the characterization of cel-
resonance (NMR) spectroscopy [165] are widely utilized lular diversity [176], the analysis of cellular heterogeneity
technologies for metabolomics analysis. Recent advance- [177], as well as the discovery of novel cell types [178].
ments have enabled the analysis of single-cell metabo- In recent years, the great advances in single-cell multi-
lomics and even at the level of single-organelle [166]. A omics technologies have enabled scientists to character-
comprehensive overview of the history and advances of ize various molecular information within individual cells,
metabolomic methodologies is provided in this review deepening our understanding of different cell types.
[167], but this review refrains from further elaboration. The largest cell-atlas initiative, the Human Cell Atlas
Importantly, it remains a challenge to combine metabo- was launched in 2017, aiming to integrate single-cell
lomics with other single-cell multi-omics technolo- omic data into comprehensive atlases, thereby enhancing
gies given the limited sample size of single cells and the our understanding of cell development, physiology, and
destructive quality of analyses and sequencing. In this CCIs [74]. Currently, with the joint efforts of scientists,
respect, advanced technologies that possess the poten- cell atlases of a wide range of organs and disease tissues
tial to integrate with other omics-based technologies are have been constructed [179, 180]. By comparing the cell
warranted. atlases of healthy and diseased states, we can uncover
the mechanisms underlying diseases and advance their
Cell-cell interactions (CCIs) diagnosis and treatment [181]. In addition, single-cell
With the advancement of bioinformatic analysis, tools multi-omics technologies can be applied to construct cell
developed based on scRNA-seq for inferring cell-cell atlases of organoids for fundamental research as dem-
communications, such as CellChat [168], CellPhoneDB onstrated in research that constructed a human brain
[13], NicheNet [169], and CellCall [170], have become organoid development atlas by co-profiling of transcrip-
mainstreams. However, these methods of inferring CCIs tome and chromatin accessibility to investigate the regu-
through algorithms are influenced by parameters and do lation of cell fate decisions [182]. Therefore, constructing
not necessarily represent actual occurrences. The appli- comprehensive cell atlases with single-cell multi-omics
cation of chemical biology tools in medical research is technologies will have profound impacts on biological
expanding, with proximity labeling technologies that may research and human health.
become game changers for reflecting real-world CCIs
[171–174]. A groundbreaking research reported that Developmental biology
FucoID, a chemical biology tool for capturing tumor anti- A principal application of single-cell multi-omics tech-
gen-specific T cells through dendritic cell interactions nologies in developmental biology is lineage tracing,
using fucosyltransferase (FT) [171]. Based on this, they which aims to track the progeny of individual cells to
further developed an advanced platform for T cell-cancer investigate cellular differentiation trajectories [183, 184].
cell and B cell-dendritic cell (DC) interactions adapted Compared with traditional methods to study cell lineage
Wu et al. Biomarker Research (2024) 12:110 Page 15 of 28
with heritable tags or naturally occurring somatic muta- of human hematopoiesis, revealing the clonal architec-
tions [185], single-cell multi-omics technologies provide ture, functional heterogeneity, and age-related changes
powerful tools for delineating comprehensive lineage of hematopoietic stem cells. Another single-cell lineage-
relationships and diverse cellular states. A newly devel- tracing technology, Camallia-seq, enables integrative
oped multi-omics technology, single-cell Regulatory analysis of chromatin accessibility, DNA methylation,
multi-omics with Deep Mitochondrial mutation profil- transcriptome, as well as cell lineage information, bring-
ing (ReDeeM), utilizes naturally occurring mitochondrial ing new insights into how cell fate decisions are regulated
DNA mutations as barcodes for lineage tracing while and how cell identities are maintained under different
analyzing transcriptome and chromatin accessibility modalities [187].
[186]. It was employed to construct a phylogenetic tree
Wu et al. Biomarker Research (2024) 12:110 Page 16 of 28
Additionally, single-cell multi-omics technologies have comprehensive gene regulatory network and elucidate
been employed for multiple stages of embryonic develop- the regulatory and causal relationships between various
ment, including preimplantation [188, 189], implantation molecules, thus holding great potential in discovering
[190], gastrulation [191], and early organogenesis [192], novel targets [200]. Olatoke et al. performed an integra-
to explore cell fate decisions of embryonic development tive scRNA-seq/single-nucleus ATAC sequencing (snA-
from multiple dimensions, thereby providing a paradigm TAC-seq) analysis on lymphangioleiomyomatosis (LAM)
to decipher the molecular programs of tissue architec- to construct a HOX-PBX gene regulatory network that
ture and cellular organization [193]. To comprehensively controlled the survival of LAM cells, thereby providing
understand embryonic development, it is essential to potential therapeutic targets for LAM [201]. Pozniak et
explore the spatial information of cells, as it is one of al. integrated single-cell transcriptomics with spatial
the key factors determining cellular identity. Integrated transcriptomics and proteomics to investigate mela-
analysis of single-cell and spatial transcriptomics can be noma, revealing a TCF4-dependent regulatory network,
applied to embryonic development, as in a study that which orchestrated multiple transcriptional programs
precisely characterized human embryonic limb develop- leading to immunotherapy resistance [202]. Targeting
ment over time and space [194]. TCF4 can enhance the sensitivity of melanoma to ICI
Altogether, with rapid advances in lineage tracing, sin- and targeted therapy. In another study, scRNA-seq and
gle-cell multi-omics, and spatial transcriptomics, we will ST were applied to characterize the cellular composition
address fundamental questions of developmental biology, and spatial structure of multiple primary lung cancers
achieving a better understanding of cell differentiation (MPLCs), finding that TNFRSF18 was highly expressed
and development. in T&NK cells within tumor tissues [203]. TNFRSF18 has
been demonstrated to be associated with non-response
Pathways identification to anti-PD-1 therapy in lung cancer [204]. Thus, it is
The currently thriving single-cell multi-omics tech- anticipated that single-cell multi-omics and spatial tech-
nologies are applied to identifying signaling pathways nologies will create a more comprehensive framework,
required for cellular function, shedding light on the providing unprecedented opportunities to discover novel
mechanisms of multiple key pathological processes targets for disease intervention.
[195–199]. Fan et al. performed an integrative analysis
of cervical squamous cell carcinoma (CSCC) utilizing Perspective in applications in clinical practice
scRNA-seq, Stereo-seq, and spatial proteomics, iden- The advent of single-cell multi-omics technologies has
tifying eight meta-programs (MP) [195]. Notably, they added breadth and depth to understanding a battery of
revealed that MP6 tumor cells interact with cancer- complex diseases and their pathology including neuro-
associated fibroblasts (CAFs) to shape an immune exclu- logical disease, immune disorders, oncology, and others.
sionary microenvironment via the FABP5-mediated Within this section, we delve into the applications of sin-
transforming growth factor β (TGFβ) pathway. Han et gle-cell multi-omics across diverse fields, underscoring
al. applied scRNA-seq and scATAC-seq to characterize its transformative impact on clinical practice.
neuroendocrine prostate cancer (NEPC) cells, identify-
ing the KIT pathway, which can be activated by FOXA2 Tumor immunology
to maintain cancer cell proliferation [196]. Inhibition Cancer therapies are continually being developed and
of KIT can be a potential strategy for the treatment of optimized, most of which can remodel the TME. As
NEPC. In addition, integrated analysis of epigenomics knowledge of the immune system improves, new immu-
and transcriptomics was applied to investigate the tran- notherapies, represented by ICIs and adoptive cell ther-
scriptional dynamics of the fibrotic kidney, revealing that apy (ACT), are emerging. The capability of single-cell
the TF Nfix could regulate the expression of the apopto- multi-omics technologies to uncover the intricate inter-
sis-related gene Ifi27 [198]. The Nfix-Ifi27 pathway was actions between the diverse cells and cancer cells sheds
also identified, which can cause kidney fibrosis by pro- light on the heterogeneity and complexity of TME, posi-
moting apoptosis. Therefore, leveraging single-cell multi- tioning them as promising tools in the exploration of
omics technologies to identify signaling pathways offers cancer treatment strategies.
crucial insights into the mechanisms of diseases, opening
up promising avenues for the development of innovative ICIs
therapeutic strategies. How do T cells react during ICI therapy? Paired scRNA-
seq and scTCR-seq might provide deep profiling. One
Novel targets discovery study from ICI therapy for NSCLC, deciphered tumor-
The single-cell multi-omics technologies can inte- specific T cell clonotype feature, regional distribution,
grate information at multiple levels to construct a and temporal persistence during ICI therapy [205].
Wu et al. Biomarker Research (2024) 12:110 Page 17 of 28
Another study from Qiu et al. utilized scRNA-seq, par- rRNA-seq and scRNA-seq analysis have been widely
alleled with scTCR/BCR-seq to elucidate the treatment utilized in microbiota gastric cancer [212], pancreatic
response of Epstein-Barr virus (EBV)-associated gas- injury [213], cholangiocarcinoma [214], etc., explor-
tric cancer. Notably, re-emerged clonotypes in ISG- ing the potential relationship between microbe and host
15+CD8+ T cells after treatment among EBV (+) patients cell types by complementing the composition of micro-
were detected and associated with effector T population bial communities and host cell and genetic informa-
expressing CXCL13 in responsive EBV (+) tumor, indi- tion. A groundbreaking study integrated 16 S rRNA and
cating their significant importance in tumor immunoche- scRNA-seq to reveal that Streptococcus anginosus pro-
motherapy response [25]. David Y. Oh et al. assessed the motes gastric tumorigenesis [215]. Jia et al. integrated
transcriptome characteristics of T cells and paired TCR 16 S rRNA-seq with single-cell transcriptomics, TCR-
from human bladder tumors. Unexpectedly, the typical seq, and ATAC-seq to reveal that IPA activates progen-
CD8+ T cell states were unchanged in tumor and normal itor-exhausted CD8+ T cells through H3K27 acetylation
tissues, while cytotoxic CD4+ T cells showed the oppo- modification [216]. It is worth mentioning that Chai et
site. They also managed to predict the therapeutic effect al. employed the Kraken method [217, 218] to process
of anti-PD-L1 in bladder cancer patients based on CD4 scRNA-seq to obtain the bacterial population corre-
signature score [206]. A study combined scRNA-seq, sponding to specific cell types [214].
TCR-seq, and ATAC-seq for integrated analysis, suggest-
ing that TdLN-TTSM cells are primary memory T cells Infectious diseases
that respond to ICI treatment, representing adoptive In the realm of infectious diseases, the technologies shed
these cells a promising immunotherapy approach [207]. light on host-pathogen dynamics, immune responses, and
advanced therapeutic strategies, especially in COVID-19
ACT [145, 146, 219–223]. Su et al. employed single-cell multi-
With the development of engineered T cells, ACT thera- omics (RNA, CITE, TCR/BCR, etc.) to observe unique
pies, represented by T cell receptor-T cell (TCR-T), Chi- dynamics in the behavior of specific CD8+ T cells during
meric antigens receptor-T cell (CAR-T), and TILs have the recuperation phase from COVID-19, among patients
reshaped the landscape of tumor treatment. Tumor- suffering from gastrointestinal sequelae [219]. Besides,
specific TCRs can recognize tumor-specific antigens, the method is of great significance for the development
which provides a solid foundation for the development of and evaluation of vaccines. Through scRNA/TCR/BCR-
TCR-T therapy. Recent research found CXCL13, CD200, seq, Peng et al. systematically profiled the immune land-
and ENTPD1 as unique markers for tumor antigen-spe- scape after vaccinating lipid nanoparticle-mRNA [146].
cific T cells using scRNA-seq and scTCR-seq. On this A study focused on the breakthrough infection and pan-
basis, developed tumor antigen-specific TCR-T cell ther- variant antivirals, and they successfully identified elite
apies have shown significant therapeutic efficacy in autol- neutralizing antibodies (nAbs) repertoire using scRNA/
ogous patient-derived xenograft (PDX) tumors [208]. BCR-seq of B cells, which showed strong neutralizing
Because of the unclear molecular mechanisms of resis- activity targeting numerous variants [145]. In addition, it
tance to CAR-T therapy in acute lymphoblastic leukemia has been found that the crosstalk of specific T cells and B
(ALL), Bai et al. integrated scRNA-seq and CITE-seq to cells following COVID-19 vaccine treatment [220, 221].
compare responders and CD19-positive relapse patients,
during which they confirmed lack of TH2 functional- Cardiovascular disease
ity might be the cause of relapse in CAR-T treatment An in-depth exploration of cardiac disease using single-
[209]. In addition, the researchers conducted a single-cell cell technologies contributing to predicting disease,
multi-omics (RNA, TCR, and CITE-seq) study in TILs therapeutic target discovery, and stratifying patients
from NSCLC patients to establish a neoantigen-targeted [224–226]. The work from Kanemaru et al. employed
T-cell signature characterized by the frequency of clono- sc-RNA-seq, single-nucleus RNA sequencing (snRNA-
types along with the levels of CD39 protein and CXCL13 seq), snATAC-seq, and spatial transcriptomics, paving
RNA. Utilizing this signature, they were able to detect the way for the anatomy and immunology of the heart
neoantigen-reactive TCRs with a success rate [210]. [227]. Delgobo et al. focused on the transgenic T cell
receptor (TCR-M) cells and myocardial infarction (MI).
Host-microbe interactions Using scRNA/TCR-seq, they elucidated TCR-M cells
A series of thrilling advancements in the interaction expressing Treg markers like Foxp3, Il2ra, and Ctla4
between the human body and microorganisms have fully and suppressed cardiac immune responses post-MI and
illustrated the protective or pathogenic effects of bacte- improved cardiac function [228]. A hypertrophy study
ria, scRNA-seq undoubtedly is the promising method applied multiple-dimensional approaches including epi-
to answer the open questions [211]. Integrated 16 S genetic and morphological analysis to the mechanism of
Wu et al. Biomarker Research (2024) 12:110 Page 18 of 28
pressure overload [229]. In addition, a study employed The challenges of single-cell multi-omics
CyTOF, scRNA-seq, and CITE-seq to decipher the Single-cell multi-omics technologies, widely applied in
immune landscapes in the plaques of atherosclerosis (AS) both biological research and clinical practice, have been
and uncover immune alterations related to clinical car- bolstered by advancements in experimental protocols
diovascular events, suggesting potential avenues for AS and data analysis, as well as by a growing consensus on
treatment [230]. their significance. Despite these improvements, they still
face hurdles that impede their widespread applications.
Neurological and brain disease These challenges and obstacles delineate the future devel-
The high complexity of brain cells requires advanced opment and trajectory of single-cell multi-omics. The fol-
single-cell multi-omics technologies to resolve the basic lowing limitations and issues must be taken into account
gene regulation both in healthy and neuropsychiat- when performing single-cell multi-omics analyses.
ric brain tissues [231]. A 2024 review comprehensively Firstly, the high cost and strict sample requirements
compiled studies on Alzheimer’s disease (AD) using of single-cell technologies discourage many researchers
transcriptomics, metabolomics, and other advanced [131]. Developing high-throughput, cost-effective, sam-
technologies, summarizing mechanisms and targets of ple-friendly, convenient single-cell multi-omics technolo-
sex differences in AD progression [232]. Notaras et al. gies is a crucial issue, particularly for the application in
performed an integrative analysis of transcriptome and clinical practice. The high cost of single-cell multi-omics
proteome on schizophrenia organoids to identify two restricts the measurement of large-scale cohorts, lead-
disease-associated factors (BRN2 and PTN). Both BRN2 ing to data that is more often utilized for discovering
and PTN promoted neurogenesis, while PTN also inhib- new insights rather than for validation, which is com-
ited apoptosis. Besides, the depletion of BRN2 and PTN monly achieved by bulk RNA sequencing [25]. Another
can lead to schizophrenia through different mechanisms challenge is the strict requirement for sample quality.
[233]. In addition, Ji et al. demonstrated that Glutamin- Fresh tissue samples are deemed appropriate for single-
ase 1 deficiency in forebrain neurons can lead to autism cell sequencing, whereas freeze-thaw samples are gen-
spectrum disorder-like behaviors by single-cell multi- erally recommended for snRNA-seq [242, 243]. Besides,
omics analysis [234]. the quantity and viability of the cell suspension are also
As mentioned earlier, the combination of single-cell important factors in obtaining high-quality single-cell
sequencing and spatial transcriptomics holds vast prom- data and detecting all cell types in the tissue.
ise for the study of brain diseases. Li et al. employed The emerging single-cell multi-omics broadens the
scRNA-seq and spatial transcriptomics to identify two multidimensionality beyond transcriptomes and raises
distinct microglial subclusters (ICAM and IPAM). ICAM more profound questions. Developing robust and
was related to ischemia, exhibiting pro-inflammatory advanced computational methods to integrate and ana-
characteristics. In contrast, IPAM, associated with the lyze multi-dimensional single-cell data is a pressing chal-
ischemic penumbra, with inflammation-alleviating and lenge to be addressed for the maturation of single-cell
neuroprotective features. Thus, they reported that target- multi-omics [7]. Importantly, these strategies need to
ing specific microglial subclusters is a promising thera- consolidate the data across diverse dimensions and man-
peutic strategy for ischemic stroke [24]. Similarly, Han age potentially significant differences in individual omics
et al. combined scRNA-seq with spatial transcriptomics data, enhancing the understanding of cellular function
to identify LGALS9-CD44 as a crucial pathway after and state. In some contexts, inconsistencies in informa-
ischemic injury. LGALS9 and CD44 exhibited opposite tion can occur among omics data, although the measure-
effects, in which upregulation of LGALS9 favored recov- ment is conducted simultaneously within the same cells
ery from post-ischemic injury, whereas knockdown of [7, 129].
CD44 diminished the therapeutic effect of LGALS9 [235]. Key information missing or mismatch due to the tech-
These applications suggest that single-cell multi-omics nology is another issue. Sequencing depth directly affects
technologies are eminently prospective for investigat- the quality of the data obtained, and choosing the appro-
ing neuropsychiatric disorders as well as brain injuries, priate sequencing depth in scRNA-seq is crucial [244].
providing unprecedented opportunities to decipher the Deeper sequencing provides a more comprehensive gene
complexity of the brain. expression profile, while insufficient sequencing depth
In addition to the applications above (Table 3), we may result in the loss of crucial information, impact-
anticipated that single-cell multi-omics and paired bioin- ing the annotation of cell types and the interpretation of
formatics tools would provide a fundamental framework their functions. Besides, some cellular heterogeneities
for the research on a variety of complex diseases or bio- may be not well presented in the transcriptome, and fail
logical processes, including autoimmune diseases [236], to define the state of the cell. High-throughput single-
aging [237], spermatogenesis [238], and others [239, 240]. cell proteomics methods may work, which are not yet
Table 3 Applications in biological research and clinical practice
Technology Diseases Source Main findings Refer-
material ence
Cell atlases construction
scRNA-seq, spatial transcriptomics / Human The study highlighted the transformative potential of single-cell and spatial genomics in understanding disease [181]
mechanisms, and diagnosing, and treating various conditions.
scRNA-seq, scATAC-seq / Human Constructing a human brain organoid development atlas to investigate the regulation of cell fate decisions. [182]
organoids
Wu et al. Biomarker Research
scRNA-seq, scTCR-seq Ovarian cancer (OC) Human A single-cell landscape of the OC ecosystem was depicted and revealed the heterogeneity of functional pheno- [241]
types and developmental origins of macrophages in tumor tissues and ascites.
Developmental biology
Camallia-seq / Mouse A new technology, Camallia-seq, enabled integrative analysis of chromatin accessibility, DNA methylation, tran- [187]
scriptome, as well as cell lineage information, bringing new insights into how cell fate decisions are regulated
and how cell identities are maintained under different modalities.
(2024) 12:110
scRNA-seq, spatial transcriptomics / Human and Precisely characterizing human embryonic limb development over time and space. [194]
mouse
Pathways identification
scRNA-seq, spatial transcriptomics, Cervical squamous cell Human MP6 tumor cells interact with CAFs to shape an immune exclusionary microenvironment via the FABP5-mediat- [195]
spatial proteomics carcinoma (CSCC) ed TGFβ pathway.
scRNA-seq, scATAC-seq Neuroendocrine pros- Human and FOXA2 is a critical driver of the adeno-to-neuroendocrine lineage transition in prostate cancer that can activate [196]
tate cancer (NEPC) mouse the KIT pathway to maintain cancer cell proliferation.
scRNA-seq, snATAC-seq Kidney Disease Human and The Nfix-Ifi27 pathway can cause kidney fibrosis by promoting apoptosis. [198]
mouse
Novel targets discovery
scRNA-seq, snATAC-seq Lymphangioleiomyo- Human and A HOX-PBX gene regulatory network controlled the survival of LAM cells. [201]
matosis (LAM) mouse
scRNA-seq, spatial transcriptomics, Metastatic melanoma Human A TCF4-dependent regulatory network orchestrated multiple transcriptional programs leading to immuno- [202]
spatial proteomics (MM) therapy resistance.
scRNA-seq, spatial transcriptomics Multiple primary lung Human TNFRSF18, associated with non-response to anti-PD-1 therapy was highly expressed in T&NK cells within MPLCs. [203]
cancers (MPLCs)
ICIs
scRNA-seq, scTCR-seq, scBCR-seq Epstein-Barr virus- Human Re-emerged clonotypes in ISG-15+CD8+ T cells after treatment among EBV (+) patients were detected and as- [25]
associated gastric sociated with effector T population expressing CXCL13 in responsive EBV (+) tumor, indicating their significant
cancer (EBV + GC) importance in tumor immunochemotherapy response.
scRNA-seq, TCR-seq Localized bladder tran- Human Identifying distinct CD4+ and CD8+ T cell states within bladder tumors and highlighting the presence of cyto- [206]
sitional cell carcinoma toxic CD4+ T cells and various CD8+ T cell subsets.
(TCC)
scRNA-seq, ACAT-seq Hepatocellular carci- Human and TdLN-TTSM cells were primary memory T cells that respond to ICI treatment, representing the adoptive transfer [207]
noma (HCC) mouse of these cells as a promising immunotherapy approach.
ACT
scRNA-seq, CITE-seq B-cell Acute Lym- Human Lack of TH2 functionality might be the cause of relapse in CAR-T treatment. [209]
phoblastic Leukemia
(B-ALL)
Page 19 of 28
Table 3 (continued)
Technology Diseases Source Main findings Refer-
Wu et al. Biomarker Research
material ence
TCR-seq, CITE-seq Non-small cell lung Human A phenotypic signature based on CD39 and CXCL13 identified neoantigen-reactive T cells in fresh NSCLC. [210]
cancer (NSCLC)
Host-microbe interactions
16 S rRNA sequencing, scRNA-seq Intrahepatic cholan- Human P. fungorum inhibited tumor growth through alanine, aspartate, and glutamate metabolism. [214]
giocarcinoma (ICC)
(2024) 12:110
16 S rRNA sequencing, scRNA-seq Gastric cancer (GC) Mouse Streptococcus anginosus promoted gastric tumorigenesis. [215]
scRNA-seq, TCR-seq Pan-cancer Human and The commensal bacterium Lactobacillus johnsonii enhanced the efficacy of ICI therapy by modulating the stem- [216]
mouse ness of CD8+ T cells.
Infectious diseases
scRNA-seq, scBCR-seq SARS-CoV-2 Human Discovery and characterization of potent neutralizing antibodies from individuals with Omicron breakthrough [145]
infections.
scRNA-seq, BCR-seq, TCR-seq SARS-CoV-2 Mouse A systematical depiction of the immune landscape after vaccinating lipid nanoparticle-mRNA. [146]
scRNA-seq, scCITE-seq, scTCR-seq, SARS-CoV-2 Human Observing unique dynamics in the behavior of specific CD8+ T cells during the recuperation phase from COVID- [219]
plasma proteomics, metabolomics 19, among patients suffering from gastrointestinal sequelae.
Cardiovascular disease
sc-RNA-seq, snRNA-seq, snATAC-seq, / Human Constructing the human cardiac landscape and paving the way for the anatomy and immunology of the heart. [227]
spatial transcriptomics
scRNA-seq, scTCR-seq Myocardial infarction Human and TCR-M cells expressing Treg markers like Foxp3, Il2ra, and Ctla4 suppressed cardiac immune responses post-MI [228]
(MI) mouse and improved cardiac function.
scRNA-seq, CITE-seq Atherosclerosis (AS) Human Deciphering the immune landscapes in the plaques of AS and uncovering immune alterations related to clinical [230]
cardiovascular events.
Neurological and brain disease
scRNA-Seq, proteomics Schizophrenia Human Identifying two disease-associated factors, BRN2 and PTN, which promoted neurogenesis, with PTN also inhib- [233]
organoids ited apoptosis.
scRNA-seq, spatial transcriptomics Ischemic stroke Mouse Two microglial subclusters, ischemic core-associated microglia (ICAM) and ischemic penumbra-associated mi- [24]
croglia (IPAM) were identified in the brains of mice subjected to middle cerebral artery occlusion (MCAO).
scRNA-seq, spatial transcriptomics Ischemic Stroke Mouse LGALS9-CD44 is a crucial pathway after ischemic injury. LGALS9 and CD44 exhibited opposite effects, in which [235]
upregulation of LGALS9 favored recovery from post-ischemic injury, whereas knockdown of CD44 diminished
the therapeutic effect of LGALS9.
Page 20 of 28
Wu et al. Biomarker Research (2024) 12:110 Page 21 of 28
established [131]. Capturing the immune repertoire also Collectively, single-cell multi-omics methods have
faces inherent technical limitations. Instances such as essentially expanded the tools to discover the rich
measuring only TRA or TRB, or erroneous matching to resources and understand the inner workings of biologi-
more than two chains, do occur [25]. Addressing how to cal processes at the single-cell level. Given that future
effectively handle these data requires more uniform rules. multi-omics studies will aid in addressing numerous bio-
Besides, there are various causes for clonal expansion, logical research and clinical practices, the technologies
including the pressure of tumor neoantigens or infec- will become the standard toolkit for studies on molecular
tious diseases. Determining whether clonal expansion is cell biology.
due to a specific factor, like tumor antigen pressure, is a
Abbreviations
topic worth exploring. This may necessitate the creation scRNA-seq Single-cell RNA sequencing
of a dedicated immune repertoire database or the use of scTCR-seq Single-cell T cell receptor sequencing
biochemistry methods to bidirectionally decode TCR and scBCR-seq Single-cell B cell receptor sequencing
scATAC-seq Single-cell assay for transposase-accessible
pMHC, among other approaches. chromatin using sequencing
Doublet refers to two cells encapsulated into one reac- TFs Transcription factors
tion volume [245], which may confound downstream PCA Principal component analysis
UMAP Uniform manifold approximation and projection
analysis, including atlas construction, DEG analysis, and t-SNE t-distributed stochastic neighbor embedding
cell trajectory inference. The emergence of the doublet DEGs Differentially expressed genes
is especially evident when a large amount of cell volume GO Gene ontology
KEGG Kyoto encyclopedia of genes and genomes
is put in. Developing a suitable means of assessing the GSVA Gene set variation analysis
removal of double cells can prevent many strange prob- SCENIC Single-cell regulatory network inference and
lems from arising [245, 246]. clustering
CCA Canonical correlation analysis
Altogether, overcoming these challenges to integrate MMN Mutual nearest neighbors
high-quality, multimodal single-cell multi-omics data is 4sU 4-thiouridine
essential. Understanding the patterns of normal tissue U Uracil
C Cytosine
function and disease progression from a single-cell per- A Adenine
spective is inseparable from these problems being solved. G Guanine
scSLAM-seq Single-cell, thiol-(SH)-linked alkylation of RNA for
metabolic labelling sequencing
Conclusions CMV Cytomegalovirus
Single-cell sequencing provides unprecedented resolu- UMI Unique molecular identifier
tion to identify multicellular connectivity and hetero- EU 5-ethynyl-uridine
EdU 5-Ethynyl-2-deoxyuridine
geneity. Multi-omics technologies have empowered EE Enteroendocrine
the scRNA-seq to break through the original limita- gRNAs Guide RNAs
tions associated with relying solely on transcriptome TME Tumor microenvironment
ISH In situ hybridization
gene expression profiles. Integrated with transcriptome, ISS In situ sequencing
genome, metabolome, proteome, TCR/BCR, epigenome, smFISH Single-molecule fluorescence ISH
etc., and broadening the axes of timescale and spatial seqFISH Sequential FISH
MERFISH Multiplexed error-robust FISH
information, multidimensional information provides RCA Rolling-circle amplification
a comprehensive snapshot of cell types and states. In RCP Rolling-circle product
the present review, we discuss the established cutting- FISSEQ Fluorescent in situ sequencing
STARmap Spatially-resolved transcript amplicon readout
edge single-cell multi-omics technologies over the past mapping
decades. Additionally, burgeoning computational biology 3D Three-dimensional
technologies are another major step toward uncovering CNS Central nervous system
ROI Regions of interest
and deciphering the secrets within the multidimensional LCM Laser capture microdissection
datasets. These bioinformatics tools link the datasets DSP GeoMx digital spatial profiler
from different modalities elucidate the function within Geo-seq Geographical position sequencing
PC Photocleavable
different cell types, and provide a wide range of informa- ICI Immune checkpoint inhibitor
tion through mathematical modeling and artificial intel- NSCLC Non-small cell lung cancer
ligence methods. From the view of clinical practice, we NGS Next-generation sequencing
ST Spatial transcriptomics
highlight the applications in tumor immunology, thera- HDST High-definition spatial transcriptomics
peutic technologies, and drug treatments. Especially for Stereo-seq Spatial enhanced resolution omics-sequencing
the discovery of new cell populations and new targets, as Decoder-seq Dendrimeric DNA coordinate barcoding design
for spatial RNA sequencing
well as evaluation and interpretation of drugs and thera- DBiT-seq Deterministic barcoding in tissue for spatial
peutics, such as PD1 and CAR-T, are elucidated. omics sequencing
Wu et al. Biomarker Research (2024) 12:110 Page 22 of 28
10. Jovic D, Liang X, Zeng H, Lin L, Xu F, Luo Y. Single-cell RNA sequencing tech- 33. Qiu Q, Hu P, Qiu X, Govek KW, Camara PG, Wu H. Massively parallel and
nologies and applications: a brief overview. Clin Transl Med. 2022;12:e694. time-resolved RNA sequencing in single cells with scNT-seq. Nat Methods.
11. Ziegenhain C, Vieth B, Parekh S, Reinius B, Guillaumet-Adkins A, Smets M, 2020;17:991–1001.
Leonhardt H, Heyn H, Hellmann I, Enard W. Comparative analysis of single-cell 34. Lin S, Yin K, Zhang Y, Lin F, Chen X, Zeng X, Guo X, Zhang H, Song J, Yang C.
RNA sequencing methods. Mol Cell. 2017;65:631–e643634. Well-TEMP-seq as a microwell-based strategy for massively parallel profiling
12. Aibar S, Gonzalez-Blas CB, Moerman T, Huynh-Thu VA, Imrichova H, Hulsel- of single-cell temporal RNA dynamics. Nat Commun. 2023;14:1272.
mans G, Rambow F, Marine JC, Geurts P, Aerts J, et al. SCENIC: single-cell 35. Matsushima W, Herzog VA, Neumann T, Gapp K, Zuber J, Ameres SL, Miska EA.
regulatory network inference and clustering. Nat Methods. 2017;14:1083–6. Sequencing cell-type-specific transcriptomes with SLAM-ITseq. Nat Protoc.
13. Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB: 2019;14:2261–78.
inferring cell-cell communication from combined expression of multi-sub- 36. Battich N, Beumer J, de Barbanson B, Krenning L, Baron CS, Tanenbaum ME,
unit ligand-receptor complexes. Nat Protoc. 2020;15:1484–506. Clevers H, van Oudenaarden A. Sequencing metabolically labeled transcripts
14. Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, in single cells reveals mRNA turnover strategies. Science. 2020;367:1151–6.
Livak KJ, Mikkelsen TS, Rinn JL. The dynamics and regulators of cell fate 37. Lu Z, Zhang M, Lee J, Sziraki A, Anderson S, Zhang Z, Xu Z, Jiang W, Ge S,
decisions are revealed by pseudotemporal ordering of single cells. Nat Nelson PT, et al. Tracking cell-type-specific temporal dynamics in human and
Biotechnol. 2014;32:381–6. mouse brains. Cell. 2023;186:4345–e43644324.
15. Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, Cahill DP, 38. Gehart H, van Es JH, Hamer K, Beumer J, Kretzschmar K, Dekkers JF, Rios A,
Nahed BV, Curry WT, Martuza RL, et al. Single-cell RNA-seq highlights intratu- Clevers H. Identification of Enteroendocrine regulators by real-time single-
moral heterogeneity in primary glioblastoma. Science. 2014;344:1396–401. cell differentiation mapping. Cell. 2019;176:1158–e11731116.
16. Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, Baglaenko Y, 39. Kirschenbaum D, Xie K, Ingelfinger F, Katzenelenbogen Y, Abadie K, Look T,
Brenner M, Loh PR, Raychaudhuri S. Fast, sensitive and accurate integration of Sheban F, Phan TS, Li B, Zwicky P et al. Time-resolved single-cell transcrip-
single-cell data with Harmony. Nat Methods. 2019;16:1289–96. tomics defines immune trajectories in glioblastoma. Cell 2023.
17. Buttner M, Miao Z, Wolf FA, Teichmann SA, Theis FJ. A test metric for assessing 40. McKenna A, Findlay GM, Gagnon JA, Horwitz MS, Schier AF, Shendure J.
single-cell RNA-seq batch correction. Nat Methods. 2019;16:43–9. Whole-organism lineage tracing by combinatorial and cumulative genome
18. Cao J, Packer JS, Ramani V, Cusanovich DA, Huynh C, Daza R, Qiu X, Lee C, editing. Science. 2016;353:aaf7907.
Furlan SN, Steemers FJ, et al. Comprehensive single-cell transcriptional profil- 41. Bowling S, Sritharan D, Osorio FG, Nguyen M, Cheung P, Rodriguez-Fraticelli
ing of a multicellular organism. Science. 2017;357:661–7. A, Patel S, Yuan WC, Fujiwara Y, Li BE, et al. An Engineered CRISPR-Cas9 Mouse
19. Gehring J, Hwee Park J, Chen S, Thomson M, Pachter L. Highly multiplexed line for simultaneous readout of lineage histories and gene expression
single-cell RNA-seq by DNA oligonucleotide tagging of cellular proteins. Nat profiles in single cells. Cell. 2020;181:1410–e14221427.
Biotechnol. 2020;38:35–8. 42. Chan MM, Smith ZD, Grosswendt S, Kretzmer H, Norman TM, Adamson B,
20. Shin D, Lee W, Lee JH, Bang D. Multiplexed single-cell RNA-seq via transient Jost M, Quinn JJ, Yang D, Jones MG, et al. Molecular recording of mammalian
barcoding for simultaneous expression profiling of various drug perturba- embryogenesis. Nature. 2019;570:77–82.
tions. Sci Adv. 2019;5:eaav2249. 43. Quinn JJ, Jones MG, Okimoto RA, Nanjo S, Chan MM, Yosef N, Bivona TG,
21. McGinnis CS, Patterson DM, Winkler J, Conrad DN, Hein MY, Srivastava V, Hu Weissman JS. Single-cell lineages reveal the rates, routes, and drivers of
JL, Murrow LM, Weissman JS, Werb Z, et al. MULTI-seq: sample multiplexing metastasis in cancer xenografts. Science 2021, 371.
for single-cell RNA sequencing using lipid-tagged indices. Nat Methods. 44. Hendriks GJ, Jung LA, Larsson AJM, Lidschreiber M, Andersson Forsman O,
2019;16:619–26. Lidschreiber K, Cramer P, Sandberg R. NASC-seq monitors RNA synthesis in
22. Zhao X, Sun S, Yu W, Zhu W, Zhao Z, Zhou Y, Ding X, Fang N, Yang R, Li JP. single cells. Nat Commun. 2019;10:3138.
Improved ClickTags enable live-cell barcoding for highly multiplexed single 45. Cao J, Zhou W, Steemers F, Trapnell C, Shendure J. Sci-fate characterizes the
cell sequencing. RSC Chem Biol. 2022;3:1052–60. dynamics of gene expression in single cells. Nat Biotechnol. 2020;38:980–8.
23. Kanton S, Boyle MJ, He Z, Santel M, Weigert A, Sanchis-Calleja F, Guijarro P, 46. Zhou Y, Jia E, Pan M, Zhao X, Ge Q. Encoding method of single-cell spatial
Sidow L, Fleck JS, Han D, et al. Organoid single-cell genomic atlas uncovers transcriptomics sequencing. Int J Biol Sci. 2020;16:2663–74.
human-specific features of brain development. Nature. 2019;574:418–22. 47. Andersson A, Larsson L, Stenbeck L, Salmén F, Ehinger A, Wu SZ, Al-Eryani G,
24. Li H, Liu P, Zhang B, Yuan Z, Guo M, Zou X, Qian Y, Deng S, Zhu L, Cao X, et Roden D, Swarbrick A, Borg Å, et al. Spatial deconvolution of HER2-positive
al. Acute ischemia induces spatially and transcriptionally distinct microglial breast cancer delineates tumor-associated cell type interactions. Nat Com-
subclusters. Genome Med. 2023;15:109. mun. 2021;12:6012.
25. Qiu MZ, Wang C, Wu Z, Zhao Q, Zhao Z, Huang CY, Wu W, Yang LQ, Zhou ZW, 48. Method of the Year. 2020: spatially resolved transcriptomics. Nat Methods
Zheng Y, et al. Dynamic single-cell mapping unveils Epstein–Barr virus- 2021, 18:1.
imprinted T-cell exhaustion and on-treatment response. Signal Transduct 49. McNicol AM, Farquharson MA. In situ hybridization and its diagnostic applica-
Target Ther. 2023;8:370. tions in pathology. J Pathol. 1997;182:250–61.
26. La Manno G, Soldatov R, Zeisel A, Braun E, Hochgerner H, Petukhov V, Lidsch- 50. Femino AM, Fay FS, Fogarty K, Singer RH. Visualization of single RNA tran-
reiber K, Kastriti ME, Lonnerberg P, Furlan A, et al. RNA velocity of single cells. scripts in situ. Science. 1998;280:585–90.
Nature. 2018;560:494–8. 51. Lubeck E, Coskun AF, Zhiyentayev T, Ahmad M, Cai L. Single-cell in situ RNA
27. Setty M, Kiseliovas V, Levine J, Gayoso A, Mazutis L. Pe’er D: characterization profiling by sequential hybridization. Nat Methods. 2014;11:360–1.
of cell fate probabilities in single-cell data with Palantir. Nat Biotechnol. 52. Williams CG, Lee HJ, Asatsuma T, Vento-Tormo R, Haque A. An introduction to
2019;37:451–60. spatial transcriptomics for biomedical research. Genome Med. 2022;14:68.
28. Gulati GS, Sikandar SS, Wesche DJ, Manjunath A, Bharadwaj A, Berger MJ, 53. Eng CL, Lawson M, Zhu Q, Dries R, Koulena N, Takei Y, Yun J, Cronin C, Karp C,
Ilagan F, Kuo AH, Hsieh RW, Cai S, et al. Single-cell transcriptional diversity is a Yuan GC, Cai L. Transcriptome-scale super-resolved imaging in tissues by RNA
hallmark of developmental potential. Science. 2020;367:405–11. seqFISH. Nature. 2019;568:235–9.
29. Guo M, Bao EL, Wagner M, Whitsett JA, Xu Y. SLICE: determining cell dif- 54. Chen KH, Boettiger AN, Moffitt JR, Wang S, Zhuang X. RNA imaging.
ferentiation and lineage based on single cell entropy. Nucleic Acids Res. Spatially resolved, highly multiplexed RNA profiling in single cells. Science.
2017;45:e54. 2015;348:aaa6090.
30. Tani H, Mizutani R, Salam KA, Tano K, Ijiri K, Wakamatsu A, Isogai T, Suzuki Y, 55. Ke R, Mignardi M, Pacureanu A, Svedlund J, Botling J, Wählby C, Nilsson M. In
Akimitsu N. Genome-wide determination of RNA stability reveals hundreds of situ sequencing for RNA analysis in preserved tissue and cells. Nat Methods.
short-lived noncoding transcripts in mammals. Genome Res. 2012;22:947–56. 2013;10:857–60.
31. Herzog VA, Reichholf B, Neumann T, Rescheneder P, Bhat P, Burkard TR, 56. Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Yang JL, Ferrante TC, Terry
Wlotzka W, von Haeseler A, Zuber J, Ameres SL. Thiol-linked alkylation of RNA R, Jeanty SS, Li C, Amamoto R, et al. Highly multiplexed subcellular RNA
to assess expression dynamics. Nat Methods. 2017;14:1198–204. sequencing in situ. Science. 2014;343:1360–3.
32. Erhard F, Baptista MAP, Krammer T, Hennig T, Lange M, Arampatzi P, Jurges CS, 57. Wang X, Allen WE, Wright MA, Sylwestrak EL, Samusik N, Vesuna S, Evans K, Liu
Theis FJ, Saliba AE, Dolken L. scSLAM-seq reveals core features of transcription C, Ramakrishnan C, Liu J et al. Three-dimensional intact-tissue sequencing of
dynamics in single cells. Nature. 2019;571:419–23. single-cell transcriptional states. Science 2018, 361.
58. Zeng H, Huang J, Zhou H, Meilandt WJ, Dejanovic B, Zhou Y, Bohlen CJ, Lee
SH, Ren J, Liu A, et al. Integrative in situ mapping of single-cell transcriptional
Wu et al. Biomarker Research (2024) 12:110 Page 24 of 28
states and tissue histopathology in a mouse model of Alzheimer’s disease. 82. Chen A, Liao S, Cheng M, Ma K, Wu L, Lai Y, Qiu X, Yang J, Xu J, Hao S, et al.
Nat Neurosci. 2023;26:430–46. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA
59. Liao J, Lu X, Shao X, Zhu L, Fan X. Uncovering an Organ’s Molecular Archi- nanoball-patterned arrays. Cell. 2022;185:1777–e17921721.
tecture at single-cell resolution by spatially resolved transcriptomics. Trends 83. Cao J, Zheng Z, Sun D, Chen X, Cheng R, Lv T, An Y, Zheng J, Song J, Wu L,
Biotechnol. 2021;39:43–58. Yang C. Decoder-Seq enhances mRNA capture efficiency in spatial RNA
60. Shi H, He Y, Zhou Y, Huang J, Maher K, Wang B, Tang Z, Luo S, Tan P, Wu M, et sequencing. Nat Biotechnol 2024.
al. Spatial atlas of the mouse central nervous system at molecular resolution. 84. Tang L. Spatially resolved multiomics. Nat Methods. 2023;20:1871.
Nature. 2023;622:552–61. 85. Liu Y, Yang M, Deng Y, Su G, Enninful A, Guo CC, Tebaldi T, Zhang D, Kim D,
61. Yao Z, van Velthoven CTJ, Kunst M, Zhang M, McMillen D, Lee C, Jung W, Bai Z, et al. High-spatial-resolution multi-omics sequencing via Deterministic
Goldy J, Abdelhak A, Aitken M, et al. A high-resolution transcriptomic and Barcoding in tissue. Cell. 2020;183:1665–e16811618.
spatial atlas of cell types in the whole mouse brain. Nature. 2023;624:317–32. 86. Zhang D, Deng Y, Kukanja P, Agirre E, Bartosovic M, Dong M, Ma C, Ma S, Su
62. Shi H, Gao L, Kirby N, Shao B, Shan X, Kudo M, Silasi R, McDaniel MJ, Zhou M, G, Bao S, et al. Spatial epigenome-transcriptome co-profiling of mammalian
McGee S et al. Clearance of VWF by hepatic macrophages is critical for the tissues. Nature. 2023;616:113–22.
protective effect of ADAMTS13 in sickle cell anemia mice. Blood 2023. 87. Liu Y, DiStasio M, Su G, Asashima H, Enninful A, Qin X, Deng Y, Nam J, Gao F,
63. Lu Y, Liu M, Yang J, Weissman SM, Pan X, Katz SG, Wang S. Spatial transcrip- Bordignon P, et al. High-plex protein and whole transcriptome co-mapping
tome profiling by MERFISH reveals fetal liver hematopoietic stem cell niche at cellular resolution with spatial CITE-seq. Nat Biotechnol. 2023;41:1405–9.
architecture. Cell Discov. 2021;7:47. 88. Russell AJC, Weir JA, Nadaf NM, Shabet M, Kumar V, Kambhampati S, Raichur
64. Lohoff T, Ghazanfar S, Missarova A, Koulena N, Pierson N, Griffiths JA, Bardot R, Marrero GJ, Liu S, Balderrama KS, et al. Slide-tags enables single-nucleus
ES, Eng CL, Tyser RCV, Argelaguet R, et al. Integration of spatial and single- barcoding for multimodal spatial genomics. Nature. 2024;625:101–9.
cell transcriptomic data elucidates mouse organogenesis. Nat Biotechnol. 89. Zhang L, Lee M, Maslov AY, Montagna C, Vijg J, Dong X. Analyzing
2022;40:74–85. somatic mutations by single-cell whole-genome sequencing. Nat Protoc.
65. Guo W, Hu Y, Qian J, Zhu L, Cheng J, Liao J, Fan X. Laser capture microdissec- 2024;19:487–516.
tion for biomedical research: towards high-throughput, multi-omics, and 90. Huang Z, Sun S, Lee M, Maslov AY, Shi M, Waldman S, Marsh A, Siddiqui T,
single-cell resolution. J Genet Genomics. 2023;50:641–51. Dong X, Peter Y, et al. Single-cell analysis of somatic mutations in human
66. Chen J, Suo S, Tam PP, Han JJ, Peng G, Jing N. Spatial transcriptomic analysis bronchial epithelial cells in relation to aging and smoking. Nat Genet.
of cryosectioned tissue samples with Geo-Seq. Nat Protoc. 2017;12:566–80. 2022;54:492–8.
67. Rosenberger FA, Thielert M, Strauss MT, Schweizer L, Ammar C, Mädler SC, 91. Zhang L, Dong X, Lee M, Maslov AY, Wang T, Vijg J. Single-cell whole-
Metousis A, Skowronek P, Wahle M, Madden K, et al. Spatial single-cell mass genome sequencing reveals the functional landscape of somatic mutations
spectrometry defines zonation of the hepatocyte proteome. Nat Methods. in B lymphocytes across the human lifespan. Proc Natl Acad Sci U S A.
2023;20:1530–6. 2019;116:9014–9.
68. Merritt CR, Ong GT, Church SE, Barker K, Danaher P, Geiss G, Hoang M, Jung 92. Cuomo ASE, Nathan A, Raychaudhuri S, MacArthur DG, Powell JE. Single-cell
J, Liang Y, McKay-Fleisch J, et al. Multiplex digital spatial profiling of proteins genomics meets human genetics. Nat Rev Genet. 2023;24:535–49.
and RNA in fixed tissue. Nat Biotechnol. 2020;38:586–99. 93. Luo J, Wu X, Cheng Y, Chen G, Wang J, Song X. Expression quantitative trait
69. Smith KD, Prince DK, Henriksen KJ, Nicosia RF, Alpers CE, Akilesh S. Digital locus studies in the era of single-cell omics. Front Genet. 2023;14:1182579.
spatial profiling of collapsing glomerulopathy. Kidney Int. 2022;101:1017–26. 94. Kang HM, Subramaniam M, Targ S, Nguyen M, Maliskova L, McCarthy E, Wan
70. Moutafi M, Martinez-Morilla S, Divakar P, Vathiotis I, Gavrielatou N, Aung E, Wong S, Byrnes L, Lanata CM, et al. Multiplexed droplet single-cell RNA-
TN, Yaghoobi V, Fernandez AI, Zugazagoitia J, Herbst RS, et al. Discovery of sequencing using natural genetic variation. Nat Biotechnol. 2018;36:89–94.
biomarkers of resistance to Immune Checkpoint Blockade in NSCLC using 95. Ding R, Wang Q, Gong L, Zhang T, Zou X, Xiong K, Liao Q, Plass M, Li L.
high-plex digital spatial profiling. J Thorac Oncol. 2022;17:991–1001. scQTLbase: an integrated human single-cell eQTL database. Nucleic Acids
71. Bressan D, Battistoni G, Hannon GJ. The dawn of spatial omics. Science. Res 2023.
2023;381:eabq4964. 96. Macaulay IC, Haerty W, Kumar P, Li YI, Hu TX, Teng MJ, Goolam M, Saurat N,
72. Ståhl PL, Salmén F, Vickovic S, Lundmark A, Navarro JF, Magnusson J, Coupland P, Shirley LM, et al. G&T-seq: parallel sequencing of single-cell
Giacomello S, Asp M, Westholm JO, Huss M, et al. Visualization and analysis genomes and transcriptomes. Nat Methods. 2015;12:519–22.
of gene expression in tissue sections by spatial transcriptomics. Science. 97. Dey SS, Kester L, Spanjaard B, Bienko M, van Oudenaarden A. Integrated
2016;353:78–82. genome and transcriptome sequencing of the same cell. Nat Biotechnol.
73. Rao A, Barkley D, França GS, Yanai I. Exploring tissue architecture using spatial 2015;33:285–9.
transcriptomics. Nature. 2021;596:211–20. 98. Kashima Y, Sakamoto Y, Kaneko K, Seki M, Suzuki Y, Suzuki A. Single-cell
74. Baysoy A, Bai Z, Satija R, Fan R. The technological landscape and applications sequencing techniques from individual to multiomics analyses. Exp Mol Med.
of single-cell multi-omics. Nat Rev Mol Cell Biol. 2023;24:695–713. 2020;52:1419–27.
75. Olaniru OE, Kadolsky U, Kannambath S, Vaikkinen H, Fung K, Dhami P, Persaud 99. Campagna D, Telatin A, Forcato C, Vitulo N, Valle G. PASS-bis: a bisulfite aligner
SJ. Single-cell transcriptomic and spatial landscapes of the developing suitable for whole methylome analysis of Illumina and SOLiD reads. Bioinfor-
human pancreas. Cell Metab. 2023;35:184–e199185. matics. 2013;29:268–70.
76. Galeano Niño JL, Wu H, LaCourse KD, Kempchinsky AG, Baryiames A, Barber 100. Otto C, Stadler PF, Hoffmann S. Fast and sensitive mapping of bisulfite-
B, Futran N, Houlton J, Sather C, Sicinska E, et al. Effect of the intratu- treated sequencing data. Bioinformatics. 2012;28:1698–704.
moral microbiota on spatial and cellular heterogeneity in cancer. Nature. 101. Guo H, Zhu P, Guo F, Li X, Wu X, Fan X, Wen L, Tang F. Profiling DNA methy-
2022;611:810–7. lome landscapes of mammalian cells with single-cell reduced-representation
77. Maynard KR, Collado-Torres L, Weber LM, Uytingco C, Barry BK, Williams SR, bisulfite sequencing. Nat Protoc. 2015;10:645–59.
Catallini JL 2nd, Tran MN, Besich Z, Tippani M, et al. Transcriptome-scale 102. Farlik M, Sheffield NC, Nuzzo A, Datlinger P, Schönegger A, Klughammer J,
spatial gene expression in the human dorsolateral prefrontal cortex. Nat Bock C. Single-cell DNA methylome sequencing and bioinformatic inference
Neurosci. 2021;24:425–36. of epigenomic cell-state dynamics. Cell Rep. 2015;10:1386–97.
78. Hasel P, Rose IVL, Sadick JS, Kim RD, Liddelow SA. Neuroinflammatory astro- 103. Angermueller C, Clark SJ, Lee HJ, Macaulay IC, Teng MJ, Hu TX, Krueger F,
cyte subtypes in the mouse brain. Nat Neurosci. 2021;24:1475–87. Smallwood S, Ponting CP, Voet T, et al. Parallel single-cell sequencing links
79. Rodriques SG, Stickels RR, Goeva A, Martin CA, Murray E, Vanderburg CR, transcriptional and epigenetic heterogeneity. Nat Methods. 2016;13:229–32.
Welch J, Chen LM, Chen F, Macosko EZ. Slide-seq: a scalable technology 104. Smallwood SA, Lee HJ, Angermueller C, Krueger F, Saadeh H, Peat J, Andrews
for measuring genome-wide expression at high spatial resolution. Science. SR, Stegle O, Reik W, Kelsey G. Single-cell genome-wide bisulfite sequencing
2019;363:1463–7. for assessing epigenetic heterogeneity. Nat Methods. 2014;11:817–20.
80. Stickels RR, Murray E, Kumar P, Li J, Marshall JL, Di Bella DJ, Arlotta P, Macosko 105. Hou Y, Guo H, Cao C, Li X, Hu B, Zhu P, Wu X, Wen L, Tang F, Huang Y, Peng J.
EZ, Chen F. Highly sensitive spatial transcriptomics at near-cellular resolution Single-cell triple omics sequencing reveals genetic, epigenetic, and transcrip-
with Slide-seqV2. Nat Biotechnol. 2021;39:313–9. tomic heterogeneity in hepatocellular carcinomas. Cell Res. 2016;26:304–19.
81. Vickovic S, Eraslan G, Salmén F, Klughammer J, Stenbeck L, Schapiro D, Äijö T, 106. Klemm SL, Shipony Z, Greenleaf WJ. Chromatin accessibility and the regula-
Bonneau R, Bergenstråhle L, Navarro JF, et al. High-definition spatial transcrip- tory epigenome. Nat Rev Genet. 2019;20:207–20.
tomics for in situ tissue profiling. Nat Methods. 2019;16:987–90.
Wu et al. Biomarker Research (2024) 12:110 Page 25 of 28
107. Simon JM, Giresi PG, Davis IJ, Lieb JD. Using formaldehyde-assisted isolation reveals it as a cell-surface Indicator of glycolytic activity of CD8(+) T cells. J
of regulatory elements (FAIRE) to isolate active regulatory DNA. Nat Protoc. Am Chem Soc. 2023;145:12701–16.
2012;7:256–67. 130. Minoshima F, Ozaki H, Odaka H, Tateno H. Integrated analysis of glycan and
108. He HH, Meyer CA, Hu SS, Chen MW, Zang C, Liu Y, Rao PK, Fei T, Xu H, Long H, RNA in single cells. iScience. 2021;24:102882.
et al. Refined DNase-seq protocol and data analysis reveals intrinsic bias in 131. Zhu C, Preissl S, Ren B. Single-cell multimodal omics: the power of many. Nat
transcription factor footprint identification. Nat Methods. 2014;11:73–8. Methods. 2020;17:11–4.
109. Pajoro A, Muiño JM, Angenent GC, Kaufmann K. Profiling nucleosome occu- 132. Wu X, Li T, Jiang R, Yang X, Guo H, Yang R. Targeting MHC-I molecules for
pancy by MNase-seq: experimental protocol and computational analysis. cancer: function, mechanism, and therapeutic prospects. Mol Cancer.
Methods Mol Biol. 2018;1675:167–81. 2023;22:194.
110. Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ. Transposition 133. Davis MM, Bjorkman PJ. T-cell antigen receptor genes and T-cell recognition.
of native chromatin for fast and sensitive epigenomic profiling of open Nature. 1988;334:395–402.
chromatin, DNA-binding proteins and nucleosome position. Nat Methods. 134. Pai JA, Satpathy AT. High-throughput and single-cell T cell receptor sequenc-
2013;10:1213–8. ing technologies. Nat Methods. 2021;18:881–92.
111. Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, Snyder MP, 135. Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, Kang B, Hu R, Huang JY,
Chang HY, Greenleaf WJ. Single-cell chromatin accessibility reveals principles Zhang Q, et al. Landscape of infiltrating T cells in Liver Cancer revealed by
of regulatory variation. Nature. 2015;523:486–90. single-cell sequencing. Cell. 2017;169:1342–e13561316.
112. Cusanovich DA, Daza R, Adey A, Pliner HA, Christiansen L, Gunderson KL, 136. Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, Gao R, Kang B, Zhang Q, Huang
Steemers FJ, Trapnell C, Shendure J. Multiplex single cell profiling of chroma- JY, et al. Lineage tracking reveals dynamic relationships of T cells in colorectal
tin accessibility by combinatorial cellular indexing. Science. 2015;348:910–4. cancer. Nature. 2018;564:268–72.
113. Xu W, Wen Y, Liang Y, Xu Q, Wang X, Jin W, Chen X. A plate-based single-cell 137. Zheng L, Qin S, Si W, Wang A, Xing B, Gao R, Ren X, Wang L, Wu X, Zhang J,
ATAC-seq workflow for fast and robust profiling of chromatin accessibility. et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science.
Nat Protoc. 2021;16:4084–107. 2021;374:abe6474.
114. Ranzoni AM, Tangherloni A, Berest I, Riva SG, Myers B, Strzelecka PM, Xu J, 138. Chen SY, Yue T, Lei Q, Guo AY. TCRdb: a comprehensive database for T-cell
Panada E, Mohorianu I, Zaugg JB, Cvejic A. Integrative single-cell RNA-Seq receptor sequences with powerful search function. Nucleic Acids Res.
and ATAC-Seq analysis of human developmental hematopoiesis. Cell Stem 2021;49:D468–74.
Cell. 2021;28:472–e487477. 139. Reynisson B, Alvarez B, Paul S, Peters B, Nielsen M. NetMHCpan-4.1 and
115. Long Z, Sun C, Tang M, Wang Y, Ma J, Yu J, Wei J, Ma J, Wang B, Xie Q, Wen J. NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by
Single-cell multiomics analysis reveals regulatory programs in clear cell renal concurrent motif deconvolution and integration of MS MHC eluted ligand
cell carcinoma. Cell Discov. 2022;8:68. data. Nucleic Acids Res. 2020;48:W449–54.
116. Wen L, Tang F. Recent advances in single-cell sequencing technologies. Precis 140. Gao YC, Gao YL, Fan YX, Zhu CY, Wei ZT, Zhou C, Chuai G, Chen QC, Zhang H,
Clin Med. 2022;5:pbac002. Liu Q. Pan-peptide Meta Learning for T-cell receptor-antigen binding recogni-
117. Nakato R, Sakata T. Methods for ChIP-seq analysis: a practical workflow and tion. Nat Mach Intell. 2023;5:236–49.
advanced applications. Methods. 2021;187:44–53. 141. Hwang JK, Alt FW, Yeap LS. Related Mechanisms of Antibody Somatic
118. Grosselin K, Durand A, Marsolier J, Poitou A, Marangoni E, Nemati F, Dahmani Hypermutation and Class Switch Recombination. Microbiol Spectr 2015,
A, Lameiras S, Reyal F, Frenoy O, et al. High-throughput single-cell ChIP-seq 3:MDNA3-0037-2014.
identifies heterogeneity of chromatin states in breast cancer. Nat Genet. 142. DeKosky BJ, Ippolito GC, Deschner RP, Lavinder JJ, Wine Y, Rawlings BM,
2019;51:1060–6. Varadarajan N, Giesecke C, Dorner T, Andrews SF, et al. High-throughput
119. Kaya-Okur HS, Wu SJ, Codomo CA, Pledger ES, Bryson TD, Henikoff JG, Ahmad sequencing of the paired human immunoglobulin heavy and light chain
K, Henikoff S. CUT&Tag for efficient epigenomic profiling of small samples repertoire. Nat Biotechnol. 2013;31:166–9.
and single cells. Nat Commun 2019, 10:1930. 143. Busse CE, Czogiel I, Braun P, Arndt PF, Wardemann H. Single-cell based high-
120. Bartosovic M, Kabbe M, Castelo-Branco G. Single-cell CUT&Tag profiles throughput sequencing of full-length immunoglobulin heavy and light chain
histone modifications and transcription factors in complex tissues. Nat genes. Eur J Immunol. 2014;44:597–603.
Biotechnol. 2021;39:825–35. 144. Hu Q, Hong Y, Qi P, Lu G, Mai X, Xu S, He X, Guo Y, Gao L, Jing Z, et al. Atlas of
121. Zhu C, Zhang Y, Li YE, Lucero J, Behrens MM, Ren B. Joint profiling of histone breast cancer infiltrated B-lymphocytes revealed by paired single-cell RNA-
modifications and transcriptome in single cells from mouse brain. Nat Meth- sequencing and antigen receptor profiling. Nat Commun. 2021;12:2186.
ods. 2021;18:283–92. 145. Guo Y, Zhang G, Yang Q, Xie X, Lu Y, Cheng X, Wang H, Liang J, Tang J, Gao Y,
122. Xiong H, Luo Y, Wang Q, Yu X, He A. Single-cell joint detection of chromatin et al. Discovery and characterization of potent pan-variant SARS-CoV-2 neu-
occupancy and transcriptome enables higher-dimensional epigenomic tralizing antibodies from individuals with omicron breakthrough infection.
reconstructions. Nat Methods. 2021;18:652–60. Nat Commun. 2023;14:3537.
123. Bennett HM, Stephenson W, Rose CM, Darmanis S. Single-cell proteomics 146. Peng L, Renauer PA, Okten A, Fang Z, Park JJ, Zhou X, Lin Q, Dong MB, Filler
enabled by next-generation sequencing or mass spectrometry. Nat Methods. R, Xiong Q, et al. Variant-specific vaccination induces systems immune
2023;20:363–74. responses and potent in vivo protection against SARS-CoV-2. Cell Rep Med.
124. Capolupo L, Khven I, Lederer AR, Mazzeo L, Glousker G, Ho S, Russo F, 2022;3:100634.
Montoya JP, Bhandari DR, Bowman AP, et al. Sphingolipids control dermal 147. Jin X, Zhou W, Luo M, Wang P, Xu Z, Ma K, Cao H, Xu C, Huang Y, Cheng R et al.
fibroblast heterogeneity. Science. 2022;376:eabh1623. Global characterization of B cell receptor repertoire in COVID-19 patients by
125. Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay single-cell V(D)J sequencing. Brief Bioinform 2021, 22.
PK, Swerdlow H, Satija R, Smibert P. Simultaneous epitope and transcriptome 148. Wensel CR, Pluznick JL, Salzberg SL, Sears CL. Next-generation sequencing:
measurement in single cells. Nat Methods. 2017;14:865–8. insights to advance clinical investigations of the microbiome. J Clin Invest
126. Mimitou EP, Cheng A, Montalbano A, Hao S, Stoeckius M, Legut M, Roush 2022, 132.
T, Herrera A, Papalexi E, Ouyang Z, et al. Multiplexed detection of proteins, 149. Wen T, Niu G, Chen T, Shen Q, Yuan J, Liu YX. The best practice for microbiome
transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat analysis using R. Protein Cell. 2023;14:713–25.
Methods. 2019;16:409–12. 150. Wang Q, Liu Z, Ma A, Li Z, Liu B, Ma Q. Computational methods and
127. Trzupek D, Dunstan M, Cutler AJ, Lee M, Godfrey L, Jarvis L, Rainbow DB, challenges in analyzing intratumoral microbiome data. Trends Microbiol.
Aschenbrenner D, Jones JL, Uhlig HH, et al. Discovery of CD80 and CD86 as 2023;31:707–22.
recent activation markers on regulatory T cells by protein-RNA single-cell 151. Jin J, Yamamoto R, Shiroguchi K. High-throughput identification and quan-
analysis. Genome Med. 2020;12:55. tification of bacterial cells in the microbiota based on 16S rRNA sequencing
128. Kearney CJ, Vervoort SJ, Ramsbottom KM, Todorovski I, Lelliott EJ, Zethoven with single-base accuracy using BarBIQ. Nat Protoc. 2024;19:207–39.
M, Pijpers L, Martin BP, Semple T, Martelotto L et al. SUGAR-seq enables 152. Kuchina A, Brettner LM, Paleologu L, Roco CM, Rosenberg AB, Carignano A,
simultaneous detection of glycans, epitopes, and the transcriptome in single Kibler R, Hirano M, DePaolo RW, Seelig G. Microbial single-cell RNA sequenc-
cells. Sci Adv 2021, 7. ing by split-pool barcoding. Science 2021, 371.
129. Yu W, Zhao X, Jalloh AS, Li Y, Zhao Y, Dinner B, Yang Y, Ouyang S, Tian T, Zhao 153. Ma P, Amemiya HM, He LL, Gandhi SJ, Nicol R, Bhattacha-
Z, et al. Chemoenzymatic Measurement of LacNAc in single-cell Multiomics ryya RP, Smillie CS, Hung DT. Bacterial droplet-based single-cell
Wu et al. Biomarker Research (2024) 12:110 Page 26 of 28
RNA-seq reveals antibiotic-associated heterogeneous cellular states. Cell. 177. Aizarani N, Saviano A, Sagar, Mailly L, Durand S, Herman JS, Pessaux P, Bau-
2023;186:877–e891814. mert TF. Grün D: A human liver cell atlas reveals heterogeneity and epithelial
154. Zheng W, Zhao S, Yin Y, Zhang H, Needham DM, Evans ED, Dai CL, Lu PJ, Alm progenitors. Nature 2019, 572:199–204.
EJ, Weitz DA. High-throughput, single-microbe genomics with strain resolu- 178. Giordani L, He GJ, Negroni E, Sakai H, Law JYC, Siu MM, Wan R, Corneau A,
tion, applied to a human gut microbiome. Science. 2022;376:eabm1483. Tajbakhsh S, Cheung TH, Le Grand F. High-dimensional single-cell cartog-
155. Li TH, Liu L, Hou YY, Shen SN, Wang TT. C-type lectin receptor-mediated raphy reveals novel skeletal muscle-resident cell populations. Mol Cell.
immune recognition and response of the microbiota in the gut. Gastroen- 2019;74:609–e621606.
terol Rep (Oxf ). 2019;7:312–21. 179. Lake BB, Menon R, Winfree S, Hu Q, Melo Ferreira R, Kalhor K, Barwinska D,
156. Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Otto EA, Ferkowicz M, Diep D, et al. An atlas of healthy and injured cell states
Huang K, Guevara F, Nelson T, et al. Analysis of Fusobacterium persistence and niches in the human kidney. Nature. 2023;619:585–94.
and antibiotic response in colorectal cancer. Science. 2017;358:1443–8. 180. Sikkema L, Ramírez-Suástegui C, Strobl DC, Gillett TE, Zappia L, Madissoon E,
157. Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, Fluckiger Markov NS, Zaragosi LE, Ji Y, Ansari M, et al. An integrated cell atlas of the lung
A, Messaoudene M, Rauber C, Roberti MP, et al. Gut microbiome influences in health and disease. Nat Med. 2023;29:1563–77.
efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 181. Rood JE, Maartens A, Hupalowska A, Teichmann SA, Regev A. Impact of the
2018;359:91–7. human cell Atlas on medicine. Nat Med. 2022;28:2486–96.
158. Galeano Nino JL, Wu H, LaCourse KD, Kempchinsky AG, Baryiames A, Barber 182. Fleck JS, Jansen SMJ, Wollny D, Zenk F, Seimiya M, Jain A, Okamoto R, Santel
B, Futran N, Houlton J, Sather C, Sicinska E, et al. Effect of the intratu- M, He Z, Camp JG, Treutlein B. Inferring and perturbing cell fate regulomes in
moral microbiota on spatial and cellular heterogeneity in cancer. Nature. human brain organoids. Nature. 2023;621:365–72.
2022;611:810–7. 183. Baron CS, van Oudenaarden A. Unravelling cellular relationships during
159. Galeano Nino JL, Wu H, LaCourse KD, Srinivasan H, Fitzgibbon M, Minot SS, development and regeneration using genetic lineage tracing. Nat Rev Mol
Sather C, Johnston CD, Bullman S. INVADEseq to identify cell-adherent or Cell Biol. 2019;20:753–65.
invasive bacteria and the associated host transcriptome at single-cell-level 184. Mukhopadhyay M. Tracing cell relationships. Nat Methods. 2022;19:27.
resolution. Nat Protoc. 2023;18:3355–89. 185. Chen C, Liao Y, Peng G. Connecting past and present: single-cell lineage trac-
160. Ghaddar B, Biswas A, Harris C, Omary MB, Carpizo DR, De Blaser MJ. Tumor ing. Protein Cell. 2022;13:790–807.
microbiome links cellular programs and immunity in pancreatic cancer. 186. Weng C, Yu F, Yang D, Poeschla M, Liggett LA, Jones MG, Qiu X, Wahlster L,
Cancer Cell. 2022;40:1240–e12531245. Caulier A, Hussmann JA et al. Deciphering cell states and genealogies of
161. Zhu H, Zou G, Wang N, Zhuang M, Xiong W, Huang G. Single-neuron iden- human haematopoiesis. Nature 2024.
tification of chemical constituents, physiological changes, and metabolism 187. Li L, Bowling S, McGeary SE, Yu Q, Lemke B, Alcedo K, Jia Y, Liu X, Ferreira
using mass spectrometry. Proc Natl Acad Sci U S A. 2017;114:2586–91. M, Klein AM, et al. A mouse model with high clonal barcode diversity for
162. Lombard-Banek C, Li J, Portero EP, Onjiko RM, Singer CD, Plotnick DO, Al joint lineage, transcriptomic, and epigenomic profiling in single cells. Cell.
Shabeeb RQ, Nemes P. In vivo subcellular Mass Spectrometry enables Proteo- 2023;186:5183–e51995122.
Metabolomic single-cell systems Biology in a chordate embryo develop- 188. Li L, Guo F, Gao Y, Ren Y, Yuan P, Yan L, Li R, Lian Y, Li J, Hu B, et al. Single-
ing to a normally behaving Tadpole (X. Laevis)*. Angew Chem Int Ed Engl. cell multi-omics sequencing of human early embryos. Nat Cell Biol.
2021;60:12852–8. 2018;20:847–58.
163. Kompauer M, Heiles S, Spengler B. Atmospheric pressure MALDI mass 189. Guo F, Li L, Li J, Wu X, Hu B, Zhu P, Wen L, Tang F. Single-cell multi-omics
spectrometry imaging of tissues and cells at 1.4-mum lateral resolution. Nat sequencing of mouse early embryos and embryonic stem cells. Cell Res.
Methods. 2017;14:90–6. 2017;27:967–88.
164. Bandura DR, Baranov VI, Ornatsky OI, Antonov A, Kinach R, Lou X, Pavlov S, 190. Zhou F, Wang R, Yuan P, Ren Y, Mao Y, Li R, Lian Y, Li J, Wen L, Yan L, et al.
Vorobiev S, Dick JE, Tanner SD. Mass cytometry: technique for real time single Reconstituting the transcriptome and DNA methylome landscapes of human
cell multitarget immunoassay based on inductively coupled plasma time-of- implantation. Nature. 2019;572:660–4.
flight mass spectrometry. Anal Chem. 2009;81:6813–22. 191. Argelaguet R, Clark SJ, Mohammed H, Stapel LC, Krueger C, Kapourani CA,
165. Larive CK, Barding GA Jr., Dinges MM. NMR spectroscopy for metabolomics Imaz-Rosshandler I, Lohoff T, Xiang Y, Hanna CW, et al. Multi-omics profiling of
and metabolic profiling. Anal Chem. 2015;87:133–46. mouse gastrulation at single-cell resolution. Nature. 2019;576:487–91.
166. Zhu H, Li Q, Liao T, Yin X, Chen Q, Wang Z, Dai M, Yi L, Ge S, Miao C, et 192. Clark SJ, Argelaguet R, Lohoff T, Krueger F, Drage D, Göttgens B, Marioni JC,
al. Metabolomic profiling of single enlarged lysosomes. Nat Methods. Nichols J, Reik W. Single-cell multi-omics profiling links dynamic DNA meth-
2021;18:788–98. ylation to cell fate decisions during mouse early organogenesis. Genome
167. Wang Z, Zhu H, Xiong W. Advances in mass spectrometry-based multi-scale Biol. 2022;23:202.
metabolomic methodologies and their applications in biological and clinical 193. Wen L, Li G, Huang T, Geng W, Pei H, Yang J, Zhu M, Zhang P, Hou R, Tian G,
investigations. Sci Bull (Beijing). 2023;68:2268–84. et al. Single-cell technologies: from research to application. Innov (Camb).
168. Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH, Myung P, 2022;3:100342.
Plikus MV, Nie Q. Inference and analysis of cell-cell communication using 194. Zhang B, He P, Lawrence JEG, Wang S, Tuck E, Williams BA, Roberts K, Klesh-
CellChat. Nat Commun. 2021;12:1088. chevnikov V, Mamanova L, Bolt L et al. A human embryonic limb cell atlas
169. Browaeys R, Saelens W, Saeys Y. NicheNet: modeling intercellular communica- resolved in space and time. Nature 2023.
tion by linking ligands to target genes. Nat Methods. 2020;17:159–62. 195. Fan J, Lu F, Qin T, Peng W, Zhuang X, Li Y, Hou X, Fang Z, Yang Y, Guo E,
170. Zhang Y, Liu T, Hu X, Wang M, Wang J, Zou B, Tan P, Cui T, Dou Y, Ning L, et al. et al. Multiomic analysis of cervical squamous cell carcinoma identifies
CellCall: integrating paired ligand-receptor and transcription factor activities cellular ecosystems with biological and clinical relevance. Nat Genet.
for cell-cell communication. Nucleic Acids Res. 2021;49:8520–34. 2023;55:2175–88.
171. Liu Z, Li JP, Chen M, Wu M, Shi Y, Li W, Teijaro JR, Wu P. Detecting Tumor 196. Han M, Li F, Zhang Y, Dai P, He J, Li Y, Zhu Y, Zheng J, Huang H, Bai F, Gao D.
Antigen-specific T cells via Interaction-Dependent Fucosyl-Biotinylation. Cell. FOXA2 drives lineage plasticity and KIT pathway activation in neuroendo-
2020;183:1117–e11331119. crine prostate cancer. Cancer Cell. 2022;40:1306–e13231308.
172. Qiu S, Li W, Deng T, Bi A, Yang Y, Jiang X, Li JP. Ru(bpy)(3) (2+) -Enabled cell- 197. Foster DS, Januszyk M, Delitto D, Yost KE, Griffin M, Guo J, Guardino N, Delitto
surface Photocatalytic Proximity labeling toward more efficient capture of AE, Chinta M, Burcham AR, et al. Multiomic analysis reveals conservation of
physically interacting cells. Angew Chem Int Ed Engl. 2023;62:e202303014. cancer-associated fibroblast phenotypes across species and tissue of origin.
173. Qiu S, Zhao Z, Wu M, Xue Q, Yang Y, Ouyang S, Li W, Zhong L, Wang W, Yang R, Cancer Cell. 2022;40:1392–e14061397.
et al. Use of intercellular proximity labeling to quantify and decipher cell-cell 198. Chen Z, Ye L, Zhu M, Xia C, Fan J, Chen H, Li Z, Mou S. Single cell multi-omics
interactions directed by diversified molecular pairs. Sci Adv. 2022;8:eadd2337. of fibrotic kidney reveal epigenetic regulation of antioxidation and apoptosis
174. Liu H, Luo H, Xue Q, Qin S, Qiu S, Liu S, Lin J, Li JP, Chen PR. Antigen-Specific within proximal tubule. Cell Mol Life Sci. 2024;81:56.
T cell detection via Photocatalytic Proximity Cell labeling (PhoXCELL). J Am 199. Wang H, Yang J, Cai Y, Zhao Y. Macrophages suppress cardiac reprogramming
Chem Soc. 2022;144:5517–26. of fibroblasts in vivo via IFN-mediated intercellular self-stimulating circuit.
175. Eisenstein M. Seven technologies to watch in 2024. Nature. 2024;625:844–8. Protein Cell 2024.
176. Ye F, Wang J, Li J, Mei Y, Guo G. Mapping cell atlases at the single-cell level. 200. Zhu Y, Ouyang Z, Du H, Wang M, Wang J, Sun H, Kong L, Xu Q, Ma H, Sun
Adv Sci (Weinh) 2023:e2305449. Y. New opportunities and challenges of natural products research: when
Wu et al. Biomarker Research (2024) 12:110 Page 27 of 28
target identification meets single-cell multiomics. Acta Pharm Sin B. cells in healthy recipients of BBIBP-CorV inactivated SARS-CoV-2 vaccine by
2022;12:4011–39. single-cell RNA sequencing. Emerg Microbes Infect. 2023;12:e2187245.
201. Olatoke T, Wagner A, Astrinidis A, Zhang EY, Guo M, Zhang AG, Mattam U, 222. Stephenson E, Reynolds G, Botting RA, Calero-Nieto FJ, Morgan MD, Tuong
Kopras EJ, Gupta N, Smith EP, et al. Single-cell multiomic analysis identifies a ZK, Bach K, Sungnak W, Worlock KB, Yoshida M, et al. Single-cell multi-omics
HOX-PBX gene network regulating the survival of lymphangioleiomyomato- analysis of the immune response in COVID-19. Nat Med. 2021;27:904–16.
sis cells. Sci Adv. 2023;9:eadf8549. 223. Xi C, Yan Z, Bai D, Zhang Y, Wang B, Han XX, Wu L, Shi X, Hu Z, Tang M et al.
202. Pozniak J, Pedri D, Landeloos E, Van Herck Y, Antoranz A, Vanwynsberghe L, Immune rebalancing at the maternal-fetal interface of maternal SARS-CoV-2
Nowosad A, Roda N, Makhzami S, Bervoets G, et al. A TCF4-dependent gene infection during early pregnancy. Protein Cell 2024.
regulatory network confers resistance to immunotherapy in melanoma. Cell. 224. Blaser MC, Kraler S, Luscher TF, Aikawa E. Multi-omics approaches to define
2024;187:166–e183125. calcific aortic valve Disease Pathogenesis. Circ Res. 2021;128:1371–97.
203. Wang Y, Chen D, Liu Y, Shi D, Duan C, Li J, Shi X, Zhang Y, Yu Z, Sun N, et al. 225. Miranda AMA, Janbandhu V, Maatz H, Kanemaru K, Cranley J, Teichmann SA,
Multidirectional characterization of cellular composition and spatial architec- Hubner N, Schneider MD, Harvey RP, Noseda M. Single-cell transcriptomics
ture in human multiple primary lung cancers. Cell Death Dis. 2023;14:462. for the assessment of cardiac disease. Nat Rev Cardiol. 2023;20:289–308.
204. Liu B, Hu X, Feng K, Gao R, Xue Z, Zhang S, Zhang Y, Corse E, Hu Y, Han W, 226. Gotthardt M, Badillo-Lisakowski V, Parikh VN, Ashley E, Furtado M, Carmo-
Zhang Z. Temporal single-cell tracing reveals clonal revival and expansion Fonseca M, Schudy S, Meder B, Grosch M, Steinmetz L, et al. Cardiac splicing
of precursor exhausted T cells during anti-PD-1 therapy in lung cancer. Nat as a diagnostic and therapeutic target. Nat Rev Cardiol. 2023;20:517–30.
Cancer. 2022;3:108–21. 227. Kanemaru K, Cranley J, Muraro D, Miranda AMA, Ho SY, Wilbrey-Clark A,
205. Pai JA, Hellmann MD, Sauter JL, Mattar M, Rizvi H, Woo HJ, Shah N, Nguyen Patrick Pett J, Polanski K, Richardson L, Litvinukova M, et al. Spatially resolved
EM, Uddin FZ, Quintanal-Villalonga A, et al. Lineage tracing reveals clonal pro- multiomics of human cardiac niches. Nature. 2023;619:801–10.
genitors and long-term persistence of tumor-specific T cells during immune 228. Delgobo M, Weiss E, Ashour D, Richter L, Popiolkowski L, Arampatzi P, Stangl
checkpoint blockade. Cancer Cell. 2023;41:776–e790777. V, Arias-Loza P, Mariotti-Ferrandiz E, Rainer PP, et al. Myocardial Milieu Favors
206. Oh DY, Kwek SS, Raju SS, Li T, McCarthy E, Chow E, Aran D, Ilano A, Pai CS, Local Differentiation of Regulatory T Cells. Circ Res. 2023;132:565–82.
Rancan C, et al. Intratumoral CD4(+) T cells mediate anti-tumor cytotoxicity in 229. Nomura S, Satoh M, Fujita T, Higo T, Sumida T, Ko T, Yamaguchi T, Tobita T,
human bladder Cancer. Cell. 2020;181:1612–e16251613. Naito AT, Ito M, et al. Cardiomyocyte gene programs encoding morphological
207. Huang Q, Wu X, Wang Z, Chen X, Wang L, Lu Y, Xiong D, Liu Q, Tian Y, Lin H, et and functional signatures in cardiac hypertrophy and failure. Nat Commun.
al. The primordial differentiation of tumor-specific memory CD8(+) T cells as 2018;9:4435.
bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell. 230. Fernandez DM, Rahman AH, Fernandez NF, Chudnovskiy A, Amir ED, Amadori
2022;185:4049–e40664025. L, Khan NS, Wong CK, Shamailova R, Hill CA, et al. Single-cell immune land-
208. He J, Xiong X, Yang H, Li D, Liu X, Li S, Liao S, Chen S, Wen X, Yu K, et al. scape of human atherosclerotic plaques. Nat Med. 2019;25:1576–88.
Defined tumor antigen-specific T cells potentiate personalized TCR-T 231. Dong X, Liu C, Dozmorov M. Review of multi-omics data resources and
cell therapy and prediction of immunotherapy response. Cell Res. integrative analysis for human brain disorders. Brief Funct Genomics.
2022;32:530–42. 2021;20:223–34.
209. Bai Z, Woodhouse S, Zhao Z, Arya R, Govek K, Kim D, Lundh S, Baysoy A, Sun 232. Lopez-Lee C, Torres ERS, Carling G, Gan L. Mechanisms of sex differences in
H, Deng Y, et al. Single-cell antigen-specific landscape of CAR T infusion prod- Alzheimer’s disease. Neuron 2024.
uct identifies determinants of CD19-positive relapse in patients with ALL. Sci 233. Notaras M, Lodhi A, Dündar F, Collier P, Sayles NM, Tilgner H, Greening D,
Adv. 2022;8:eabj2820. Colak D. Schizophrenia is defined by cell-specific neuropathology and
210. Hanada KI, Zhao C, Gil-Hoyos R, Gartner JJ, Chow-Parmer C, Lowery FJ, Krishna multiple neurodevelopmental mechanisms in patient-derived cerebral
S, Prickett TD, Kivitz S, Parkhurst MR, et al. A phenotypic signature that identi- organoids. Mol Psychiatry. 2022;27:1416–34.
fies neoantigen-reactive T cells in fresh human lung cancers. Cancer Cell. 234. Ji C, Tang Y, Zhang Y, Huang X, Li C, Yang Y, Wu Q, Xia X, Cai Q, Qi XR, Zheng
2022;40:479–e493476. JC. Glutaminase 1 deficiency confined in forebrain neurons causes autism
211. Westermann AJ, Vogel J. Cross-species RNA-seq for deciphering host-microbe spectrum disorder-like behaviors. Cell Rep. 2023;42:112712.
interactions. Nat Rev Genet. 2021;22:361–78. 235. Han B, Zhou S, Zhang Y, Chen S, Xi W, Liu C, Zhou X, Yuan M, Yu X, Li L, et
212. Peng R, Liu S, You W, Huang Y, Hu C, Gao Y, Jia X, Li G, Xu Z, Chen Y. Gastric al. Integrating spatial and single-cell transcriptomics to characterize the
microbiome alterations are Associated with decreased CD8 + tissue-Resident molecular and cellular architecture of the ischemic mouse brain. Sci Transl
memory T cells in the Tumor Microenvironment of Gastric Cancer. Cancer Med. 2024;16:eadg1323.
Immunol Res. 2022;10:1224–40. 236. Wang Y, Chen Z, Wang T, Guo H, Liu Y, Dang N, Hu S, Wu L, Zhang C, Ye K, Shi
213. Li G, Liu L, Lu T, Sui Y, Zhang C, Wang Y, Zhang T, Xie Y, Xiao P, Zhao Z, et al. B. A novel CD4 + CTL subtype characterized by chemotaxis and inflammation
Gut microbiota aggravates neutrophil extracellular traps-induced pancreatic is involved in the pathogenesis of Graves’ orbitopathy. Cell Mol Immunol.
injury in hypertriglyceridemic pancreatitis. Nat Commun. 2023;14:6179. 2021;18:735–45.
214. Chai X, Wang J, Li H, Gao C, Li S, Wei C, Huang J, Tian Y, Yuan J, Lu J, et al. 237. Terekhova M, Swain A, Bohacova P, Aladyeva E, Arthur L, Laha A, Mogile-
Intratumor microbiome features reveal antitumor potentials of intrahepatic nko DA, Burdess S, Sukhov V, Kleverov D, et al. Single-cell atlas of healthy
cholangiocarcinoma. Gut Microbes. 2023;15:2156255. human blood unveils age-related loss of NKG2C(+)GZMB(-)CD8(+)
215. Fu K, Cheung AHK, Wong CC, Liu W, Zhou Y, Wang F, Huang P, Yuan K, Coker memory T cells and accumulation of type 2 memory T cells. Immunity.
OO, Pan Y et al. Streptococcus anginosus promotes gastric inflammation, 2023;56:2836–e28542839.
atrophy, and tumorigenesis in mice. Cell 2024. 238. Huang Y, Li L, An G, Yang X, Cui M, Song X, Lin J, Zhang X, Yao Z, Wan C, et
216. Jia D, Wang Q, Qi Y, Jiang Y, He J, Lin Y, Sun Y, Xu J, Chen W, Fan L et al. al. Single-cell multi-omics sequencing of human spermatogenesis reveals a
Microbial metabolite enhances immunotherapy efficacy by modulating T cell DNA demethylation event associated with male meiotic recombination. Nat
stemness in pan-cancer. Cell 2024. Cell Biol. 2023;25:1520–34.
217. Wood DE, Salzberg SL. Kraken: ultrafast metagenomic sequence classification 239. Wang G, Chiou J, Zeng C, Miller M, Matta I, Han JY, Kadakia N, Okino ML,
using exact alignments. Genome Biol. 2014;15:R46. Beebe E, Mallick M, et al. Integrating genetics with single-cell multiomic
218. Lu J, Rincon N, Wood DE, Breitwieser FP, Pockrandt C, Langmead B, Salzberg measurements across disease states identifies mechanisms of beta cell
SL, Steinegger M. Metagenome analysis using the Kraken software suite. Nat dysfunction in type 2 diabetes. Nat Genet. 2023;55:984–94.
Protoc. 2022;17:2815–39. 240. Cortés-López M, Chamely P, Hawkins AG, Stanley RF, Swett AD, Ganesan
219. Su Y, Yuan D, Chen DG, Ng RH, Wang K, Choi J, Li S, Hong S, Zhang R, Xie J, S, Mouhieddine TH, Dai X, Kluegel L, Chen C, et al. Single-cell multi-omics
et al. Multiple early factors anticipate post-acute COVID-19 sequelae. Cell. defines the cell-type-specific impact of splicing aberrations in human hema-
2022;185:881–e895820. topoietic clonal outgrowths. Cell Stem Cell. 2023;30:1262–e12811268.
220. Marzano P, Balin S, Terzoli S, Della Bella S, Cazzetta V, Piazza R, Sandrock I, 241. Zheng X, Wang X, Cheng X, Liu Z, Yin Y, Li X, Huang Z, Wang Z, Guo W,
Ravens S, Tan L, Prinz I, et al. Transcriptomic profile of TNF(high) MAIT cells is Ginhoux F, et al. Single-cell analyses implicate ascites in remodeling the
linked to B cell response following SARS-CoV-2 vaccination. Front Immunol. ecosystems of primary and metastatic tumors in ovarian cancer. Nat Cancer.
2023;14:1208662. 2023;4:1138–56.
221. Tong R, Luo L, Zhao Y, Sun M, Li R, Zhong J, Chen Y, Hu L, Li Z, Shi J, et al. 242. Guo Y, Wang W, Ye K, He L, Ge Q, Huang Y, Zhao X. Single-nucleus RNA-Seq:
Characterizing the cellular and molecular variabilities of peripheral immune open the era of great Navigation for FFPE tissue. Int J Mol Sci 2023, 24.
Wu et al. Biomarker Research (2024) 12:110 Page 28 of 28
243. Bakken TE, Hodge RD, Miller JA, Yao Z, Nguyen TN, Aevermann B, Barkan 246. Sun B, Bugarin-Estrada E, Overend LE, Walker CE, Tucci FA, Bashford-Rogers
E, Bertagnolli D, Casper T, Dee N, et al. Single-nucleus and single-cell RJM. Double-jeopardy: scRNA-seq doublet/multiplet detection using multi-
transcriptomes compared in matched cortical cell types. PLoS ONE. omic profiling. Cell Rep Methods. 2021;1:None.
2018;13:e0209648.
244. Zhang MJ, Ntranos V, Tse D. Determining sequencing depth in a single-cell
RNA-seq experiment. Nat Commun. 2020;11:774. Publisher’s note
245. Xi NM, Li JJ. Benchmarking Computational Doublet-Detection methods for Springer Nature remains neutral with regard to jurisdictional claims in
single-cell RNA sequencing data. Cell Syst. 2021;12:176–e194176. published maps and institutional affiliations.