Abstract
The heterogeneity of the hematopoietic system was largely veiled by traditional bulk sequencing methods, which measure the averaged signals from mixed cellular populations. In contrast, single-cell sequencing has enabled the direct measurement of individual signals from each cell, significantly enhancing our ability to unveil such heterogeneity. Building on these advances, numerous single-cell multi-omics techniques have been developed into high-throughput, routinely accessible platforms, delineating the precise relationships among different layers of the central dogma in molecular biology. These technologies have uncovered the intricate landscape of genetic clonality and transcriptional heterogeneity in both normal and malignant hematopoietic systems, highlighting their roles in differentiation, disease progression, and therapy resistance. This review aims to provide a brief overview of the principles of single-cell technologies, their historical development, and a subset of ever-expanding multi-omics tools, emphasizing the specific research questions that inspired their creation. Amidst the evolving landscape of single-cell multi-omics technologies, our main objective is to guide investigators in selecting the most suitable platforms for their research needs.
Introduction
The hematopoietic system is a complex ecosystem comprising diverse cellular populations, each defined by different lineages, functions, and activation states. While cost-effective, the conventional approach of analyzing bulk blood specimens obscures detailed examination of heterogeneous cellular states. As distinct cellular populations are sequenced together, bulk sequencing yields an average mutant allele frequency or gene expression profile combined for the heterogeneous populations, masking the precise lineage or clonal relationship between the cells. In contrast, single-cell sequencing (SCS) has enabled direct assessment of cell-to-cell variabilities, reconstruction of evolutionary relationships, and identification of rare populations.
Historically, the hematopoietic system has been at the forefront of technological advancements due to the accessibility of blood samples and the facility of analyzing cells that naturally exist in dissociated states. Over the past decade, SCS has continued to refine our understanding of the hematopoietic system, challenging the models of hematopoiesis1 and characterizing unconventional leukemic stem cells2 conferring resistance against targeted therapies, just to iterate a few.
This review aims to summarize the principles of SCS while outlining recent advancements in SCS. As the field rapidly evolves and new technologies continue to emerge, investigators are left with an almost overwhelming abundance of choices. The primary focus of this review is to present a subset of the many available multi-omics platforms, particularly in the context of studying tumor heterogeneity and clonal evolution, while highlighting the types of research questions they are designed to address.
Overview of single-cell technologies
Single-cell isolation and library preparation techniques
The first crucial step in SCS is the isolation of individual cells. Initially, single cells were manually picked under a microscope,3 which was laborious and inherently low in throughput. Shortly after, the procedure was scaled up and partially automated using fluorescence-activated cell sorting,4,5 which was used to place each cell into each well of 96-well plates before researchers carried out library preparation.
The advancement of SCS gained considerable momentum with the introduction of microfluidics, which allowed a large number of cells to be automatically isolated and prepared for libraries in parallel. The earlier plate-based microfluidics, such as Fluidigm’s C1 system,6 enabled the processing of hundreds of cells per sample. Later, the advent of droplet-based microfluidics7,8 scaled up the throughput to thousands of cells per sample, enhancing the ability to detect rare populations in the sample. Perhaps the most well-known droplet-based platform, the Chromium from 10X Genomics,9 quickly gained popularity over existing droplet-based methods7,8 by significantly improving cell capture rates.
A significant drawback of microfluidics is their propensity to isolate multiple cells together, known as multiplets. To circumvent this, some single-cell studies adopted a high-throughput technique called combinatorial indexing,10 which reduces the likelihood of any two cells receiving the same barcode without necessitating each cell’s physical isolation. Others sought to capture single cells in scalable, size-adjusted nanowells,11 minimizing the probability of isolating more than one cell per well. Nevertheless, a comparative study12 found no significant difference in multiplet rates between these methods and microfluidics, suggesting that factors such as target sensitivity or ease of use should be prioritized when choosing high-throughput techniques. A fundamental library preparation technique in SCS involves labeling the fragmented genetic molecules with a cell-specific barcode to trace the molecules’ cell of origin. This is because single cells are typically pooled prior to sequencing to reduce costs and enhance sequencing efficiency. In addition, each molecule is commonly assigned a unique molecular identifier (UMI),¹³ which enables accurate quantification of the molecule’s original abundance before amplification. Notably, the incorporation of UMI has been instrumental in advancing SCS technologies by allowing researchers to detect and correct artifacts introduced during the aggressive amplification process. Such amplification is required for SCS because each mammalian cell contains only a few picograms of DNA or RNA, whereas sequencers require at least several nanograms of input material. Another common practice involves tagging each molecule with a sample-specific barcode, enabling researchers to pool multiple samples for sequencing and further reduce costs. Tools such as cell hashing14 directly assign sample-specific barcodes to molecules, while computational methods like demuxlet15 utilize natural markers such as single-nucleotide variants to distinguish samples based on donor-specific mutational profiles. Despite these advances, challenges such as cross-contamination persist, underscoring the need for further technical and bioinformatic improvements in sample multiplexing techniques.
Single-cell RNA sequencing
The human transcriptome, comprising approximately 20,000 genes, represents only 2% of the entire genome. This has uniquely positioned single-cell RNA sequencing (scRNA-seq) as a leading single-cell technology and a powerful, unbiased tool for capturing a cell’s phenotypic state.
The inaugural single-cell study in 20093 (Figure 1) conducted whole-transcriptome sequencing on just a single mouse blastomere. Since then, a wide spectrum of scRNA-seq protocols has been developed to meet different needs (Table 1). Given that the average mRNA transcripts are several kilobases long, reading only 100-400 base pairs at either the 3’ or 5’ end of mRNA provides a cost-effective and high-throughput approach. However, this approach cannot profile RNA isoforms or many single-nucleotide variants, which require full-length sequencing. Smart-seq2,16 a pioneer of modern full-length sequencing, has been widely used in applications that require highly sensitive variant analysis but have lower demands on the throughput. More recently, it was updated as Smart-seq3,17 incorporating 5’ unique molecular identifiers to enhance the accuracy of transcript quantification. Extending from these capabilities, recent advancements in scRNA-seq have made it compatible with long-read sequencing, such as Nanopore,18 which circumvents the error-prone processes of fragmentation and assembly. Although long-read sequencing has facilitated the study of larger structural variants in scRNA-seq, its current high error rates warrant careful application. The per-cell cost increases progressively from 3’/5’ end-based sequencing to full-length sequencing and further to long-read sequencing. While 5’ end sequencing is slightly more expensive than 3’ end sequencing, it offers improved mutation detection. These differences in costs present a unique trade-off between transcript coverage and throughput (Figure 2A), underscoring the importance of selecting the appropriate strategy based on specific experimental needs. One of the current limitations of scRNA-seq is transcriptomic dropouts. Most mRNA are expressed in only a few copies per cell, and the expression of mRNA at any given state varies significantly depending on the cell physiology and RNA stochasticity. This makes it difficult to discern whether the observed absence of a transcript is due to technical error or genuine biological variability. However, newer algorithms, as well as the construction of reference-level transcriptomic cell atlases, are gradually addressing these challenges.
Single-cell DNA sequencing
Single-cell DNA sequencing (scDNA-seq) technologies present a unique opportunity to analyze the clonality of individual cells and the order in which mutations arise, both of which have significant implications for clinical outcomes.19 However, the development of scDNA-seq was relatively delayed compared to that of scRNA-seq (Figure 1), primarily due to the limited copy number of DNA in a cell (only two versus multiple copies of mRNA), as well as the DNA’s larger size and complexity (Figure 2). These attributes of the DNA create higher risks of misalignment, allele dropout, and artifact mutations, all of which can easily complicate scDNA-seq analysis.
Table 1.Summary of single-cell sequencing technologies introduced in this review.
Compared to the transcriptome, the human genome spans several gigabases. Subsequently, whole-genome amplification remains a bottleneck in scDNA-seq, making single-cell whole-genome sequencing costly, error-prone, and challenging. To reduce the whole-genome amplification bottleneck, specific methods have emerged for different intentions of analysis. In general, polymerase chain reaction (PCR)-based methods, such as degenerate oligonucleotide-primed PCR20 or multiple annealing and looping-based amplification cycles,21 are considered more suitable for analyzing larger changes in the chromosome, such as copy number alterations. Meanwhile, isothermal methods utilizing high-fidelity phi29 polymerases, such as multiple displacement amplification22 or primary template-directed amplification,23 are deemed more suitable for precisely analyzing smaller changes in the genome, such as single-nucleotide variants. The range of options available for whole-genome amplification stands in contrast to those commonly employed for whole-transcriptome amplification in scRNA-seq, which are simply narrowed down to either in vitro transcription24 or PCR. The first scDNA-seq study in 2011 performed whole-genome sequencing on a hundred single nuclei from human breast cancer5 (Figure 1). Using degenerate oligonucleotide-primed PCR, the study focused on profiling copy number alterations to reconstruct the clonal history of breast cancer at the chromosomal level. Subsequent studies demonstrated the feasibility of single-cell whole-exome sequencing in human essential thrombocytopenia25 and renal cell carcinoma.26 Using multiple displacement amplification to profile single-nucleotide variants, these studies aimed to characterize the clonal makeup of these diseases at the single nucleotide level.
To minimize technical artifacts from in vitro whole-genome amplification, some researchers sought to utilize amplification methods naturally employed by cells. One group27 performed ex vivo whole-genome amplification via a method known as single-cell cloning, deriving colonies from individual hematopoietic stem and progenitor cells that are capable of forming colonies. This approach has found numerous applications in studies investigating the clonal architecture of hematopoietic stem and progenitor cells, such as in healthy individuals28,29 and patients with myeloproliferative neoplasms.30 Another group captured nuclei undergoing the G2/M phase of the cell cycle, leveraging their duplicated genomic material.31
The large size of the genome presents a distinct tradeoff between genome coverage and throughput in scDNA-seq (Figure 2B). Targeted scDNA-seq32,33 only requires a small region of DNA amplification but provides an incomplete picture of the genome, trading coverage for cost and efficiency. To give a practical example within commercial programs, Mission Bio’s Tapestri platform32 uses targeted scDNA-seq to profile thousands of cells at the expense of sequencing only tens or hundreds of genes. In contrast, Bioskryb’s ResolveDNA platform,34 which relies on the primary template-directed amplification technique for whole-genome amplification, provides whole-genome or whole-exome analyses for sequencing just a few hundred cells. At the single-cell level, whole-exome sequencing is done by enriching the exome region after performing whole-genome amplification (Figure 2B), facing the same bottleneck as whole-genome sequencing.
Figure 1.The milestones of single-cell sequencing platforms in the past decade. Color coding is as follows: green for single-cell RNA sequencing, orange for single-cell DNA sequencing, purple for single-cell epigenomics sequencing, red for single-cell multi-omics sequencing, pink for single-cell isolation and library preparation techniques, and turquoise for whole-genome amplification methods. DOP-PCR: degenerate oligonucleotide-primed polymerase chain reaction; MDA: multiple displacement amplification; scRNA-seq: single-cell RNA sequencing; FACS: fluorescence-activated cell sorting; scDNA-seq: single-cell DNA sequencing; MALBAC: multiple annealing and looping-based amplification cycles: scATAC-seq: single-cell assay for transposase-accessible chromatin sequencing; scTrio-seq: single-cell triple-omics sequencing; scRRBS: single-cell reduced-representation bisulfite sequencing; UMI: unique molecular identifier; CITE-seq: cellular indexing of transcriptomes and epitopes by sequencing; GoT: genotyping of transcriptomes; SPARC: single-cell protein and RNA co-profiling; PTA: primary template-directed amplification; MAESTER: mitochondrial alteration enrichment from single-cell transcriptomes to establish relatedness; GoT-ChA: genotyping of targeted loci with single-cell chromatin accessibility; ReDeeM: regulatory multimaps with deep mitochondrial mutation profiling.
One of the core applications of scDNA-seq is to trace the evolutionary trajectory through examination of mutation co-occurrence. In this sense, targeted scDNA-seq offers an efficient means for clonal analysis by focusing on common driver mutations. However, due to the limited number of mutations profiled for each cell, the resolution of clonality is inherently limited. Albeit costly, whole-genome analysis is necessary to obtain a complete and unbiased view of the genome.
Single-cell mitochondrial sequencing
To address the limitations of high cost, error rates, and lack of scalability in single-cell whole-genome sequencing, recent studies have also assessed the potential of analyzing clonality through profiling the mitochondrial DNA. A landmark study35 reported that mitochondrial DNA has proven to be a reliable source for lineage tracing due to its smaller size (16.6 kilobases) and higher copy number per cell. Most importantly, as the mitochondrial genotype is already, and often inadvertently, captured during routine scRNA-seq or single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq)36 experiments, this approach is spearheading advancements in single-cell lineage tracing techniques. However, it is currently unknown whether the clonalities inferred from mitochondrial DNA match those inferred from genomic DNA.
Bioinformatics
With the increasing standardization of experimental workflows, data analysis now constitutes the majority of the effort in single-cell analysis. Repurposing many tools used for processing bulk sequencing data, the upstream preprocessing, such as trimming, alignment, and variant calling, has become relatively well-defined and automated. For instance, many commercial SCS platforms provide cloudbased interfaces that allow users to simply upload FASTQ files and run all the steps of preprocessing. Interestingly, a study37 has even demonstrated that the choice of scRNA-seq preprocessing pipelines has minimal impact on downstream clustering results once effective normalization and clustering methods are applied.
Figure 2.The tradeoff between coverage and throughput due to the high cost of single-cell sequencing. (A) The tradeoff between cellular throughput and transcript coverage in single-cell RNA sequencing requires a choice between high-throughput sequencing with partial transcript coverage on thousands of cells and low-throughput sequencing with full transcript coverage on tens or hundreds of cells. (B) The tradeoff between cellular throughput and genome coverage in single-cell DNA sequencing requires a choice between narrow, targeted sequencing on thousands of cells and broad, genome-wide sequencing on tens or hundreds of cells. scRNA-seq: single-cell RNA sequencing; scDNA-seq: single-cell DNA sequencing.
In contrast, downstream analysis requires advanced programming skills and domain knowledge, presenting a hurdle that often necessitates hiring a trained bioinformatician to manage the analysis. The analytic workflow of scRNA-seq has been well-established, supported by publications such as ‘Best Practices of Single-Cell Analysis’38 and renowned tools such as Seurat39 or SCANPY.40 Conversely, scDNA-seq has yet to achieve a comparable level of establishment, although new tools such as Mission Bio’s Mosaic or Bioskryb’s BaseJumper are beginning to make the analysis more accessible.
Multimodal single-cell technologies
The maturation of unimodal single-cell technologies soon motivated researchers to overlay different modalities to understand relationships among various components of the cell, such as the genome, epigenome, transcriptome, or proteome (Table 1, Figure 3). Some of these technologies are bona fide multi-omics, directly measuring multiple elements from the same cell, while others attempt to infer unmeasured modalities based on a measured modality.
Integrating genomics with transcriptomics
Although scRNA-seq effectively resolves granular cell types within a tumor sample, it poses challenges in precisely demarcating leukemic cells from normal cells in the sample. In this sense, concurrent examination of a cell’s mutational status enables the assessment of malignancy in a sample of transcriptionally heterogeneous cells. This approach offers a unique advantage in discovering the underlying transcriptional pathways that may drive the predominance of certain clonal populations and elucidating the impact of mutations in the alteration of transcriptional activities. Several platforms have enabled direct measurement of DNA and RNA from the same cell. For instance, Resolve-OME,34 an extension of the ResolveDNA platform, combines whole-genome sequencing with full-length whole-transcriptome sequencing to allow for unbiased screening of a cell’s genome and transcriptome. To focus instead on only a few genes of interest, TARGET-seq41 could be used, as demonstrated in a recent study42 that investigated two representative clonal hematopoiesis mutations in DNMT3A and TET2. Using TARGET-seq, the study demonstrated that hematopoietic stem cells harboring these mutations gain a fitness advantage over wild-type hematopoietic stem cells by attenuating inflammation-associated transcriptional programs, which enhances the survival of mutant hematopoietic stem cells in the inflammatory tumor environment. However, direct measurement of DNA remains expensive and technically challenging, prompting the development of various alternatives. Inferring copy number alterations from scRNA-seq data is relatively straightforward and can be achieved using simple estimation43 or advanced computational algorithms.44 On the contrary, detecting oncogenic single-nucleotide variants and indels proves to be more challenging. Truncating or non-sense mutations are less likely to be transcribed, and 3’ or 5’ end biased scRNA-seq may not cover the mutated loci efficiently. Nonetheless, several groups have successfully inferred specific driver mutations from scRNA-seq reads by experimentally modifying the scRNA-seq protocols, such as spiking in mutant-specific primers,45,46 circularizing cDNA,46 or utilizing long-read sequencing.46,47 A notable multi-omic platform adopting this approach is genotyping of transcriptomes (GoT),46 which was used to analyze patients with essential thrombocytopenia and provided in situ evidence that the transcriptional outcome of CALR mutations may confer a clonal advantage specifically in the compartment of megakaryocyte progenitors.
Figure 3.Summary of capabilities across multi-modal single-cell platforms. GoT: genotyping of transcriptomes; GoT-ChA: genotyping of targeted loci with single-cell chromatin accessibility; scTrio-seq: single-cell triple-omics sequencing; Smart-RRBS: Smart-seq2 and reduced representation bisulfite sequencing; DAb-seq: DNA-antibody sequencing; CITE-seq: cellular indexing of transcriptomes and epitopes by sequencing; SPARC: single-cell protein and RNA co-profiling; MAESTER: mitochondrial alteration enrichment from single-cell transcriptomes to establish relatedness; ReDeeM: regulatory multimaps with deep mitochondrial mutation profiling.
Integrating genomics with epigenomics
Following the development of scRNA-seq and scDNA-seq, newer technologies were rapidly developed to study epigenomic processes at the single-cell level (Figure 1). For instance, scATAC-seq profiles open chromatin regions of the DNA, and single-cell reduced-representation bisulfite sequencing (scRRBS)48 analyzes DNA methylation patterns in the CpG-rich regions. These technologies were quickly integrated with genomic analysis to understand how alterations in gene regulation are affected by mutations at the DNA level. Two multi-omic protocols have enabled high-throughput analysis of a few target genes with chromatin accessibility by adding specific DNA primers onto scATAC-seq49,50 protocols. For example, genotyping of targeted loci with single-cell chromatin accessibility (GoT-ChA),50 an extension of a droplet-based scATAC-seq protocol, was applied to study patients with myeloproliferative neoplasm. Focusing on JAK2 V617F mutations, the study revealed epigenetic rewiring specifically in hematopoietic stem cells and megakaryocyte progenitors, marked by increased chromatin accessibility in inflammation-related genes. A notable aspect of GoT-ChA is that, beyond the two directly sequenced modalities, it sought computational integration with other multi-omic tools, such as DOGMA-seq,51 through the common grounds of an ATAC-seq modality. Hence, GoT-ChA was able to infer modalities such as mitochondrial DNA, immunophenotype, and gene expression, which were not directly sequenced by the platform itself. Such an imputation approach has become increasingly popular to complement missing information and cross-validate the findings that have been made based on measured modalities, overcoming current limitations in multi-omic capabilities that can directly sequence only two to three modalities.
Currently, multi-omics technologies that integrate DNA mutations and methylation status are rooted in joint methylome and transcriptomic analysis. Earlier methods such as the single-cell triple-omics sequencing technique (scTrio-seq)52 enabled genome, methylome, and transcriptome analysis by combining scRRBS with scRNA-seq and inferring copy number alterations from their reads. More recent methods attempted to integrate scRRBS with Smart-seq2 (Smart-RRBS)53 and, further, with genotyping54 to profile the DNA methylome, transcriptome, and single-nucleotide variants simultaneously. Smart-RRBS was applied in a study of chronic lymphocytic leukemia54 that aimed to reconstruct the evolutionary history of tumor cells based on “epimutations”, the heritable changes in DNA methylation. The study initially constructed the chronic lymphocytic leukemia lineage tree based on scRRBS data and subsequently integrated genotyping data to validate and refine the lineage tree.
Integrating genomics with proteomics
Prior to the development of scRNA-seq, cell types were traditionally identified based on surface proteins or immunophenotypes. Integrating single-cell immunophenotypes offers several advantages over single-cell transcriptomics for characterizing a cell’s phenotypic state. Not only is immunophenotype the clinically established gold standard for cell type identification, but it also exhibits greater stability compared to RNA species, holding greater potential for applications such as in vitro diagnostic tools. Moreover, certain immunophenotypes serve as canonical markers for leukemic cells, enabling their use as surrogates of malignancy to demarcate leukemic cells within a sample.
To remain compatible with high-throughput droplet-based microfluidic platforms, standard flow cytometry has been modified by conjugating protein-binding antibodies with oligonucleotides instead of fluorophores.55 This strategy allows protein abundance to be sequenced and quantified just like DNA. Another benefit of using oligonucleotides is their ability to generate nearly infinite combinations of barcodes. In practice, this allows the unique identification of hundreds of surface proteins, far exceeding the capabilities of conventional mass or flow cytometry, which can typically trace only a few dozen. However, antibody-oligonucleotide tags are not fully optimized yet, with efforts to refine antibody titration56 only recently underway.
Building on targeted scDNA-seq platforms such as Tapestri, DNA-antibody sequencing (DAb-seq)32 integrates the analysis of surface proteins and target genes to elucidate the relationship between cellular phenotypes and driver mutations. In acute myeloid leukemia research, DAb-seq has been employed to map the mutational landscape across diverse acute myeloid leukemia cell types,57,58 outline genotype-phenotype evolution in response to targeted therapies,59 and distinguish non-malignant clonal hematopoiesis mutations from common driver mutations by identifying the absence of leukemia-associated immunophenotypes within the same cell.60
Integrating transcriptomics with proteomics
Several multi-omic platforms, including the widely-used cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq),61 have been developed to measure mRNA expression and surface protein abundance from the same cell. Integrating scRNA-seq with immunophenotype data improves cell type identification by providing a more comprehensive view of a cell’s phenotypic state. For instance, a recent study56 using CITE-seq produced a high-resolution atlas of human hematopoietic progenitors, identifying novel surface markers to isolate transitional cell populations. These isolated progenitors exhibited restricted clonal outputs in experimental data, supporting a discrete stem and progenitor cell state model1 rather than the continuous hematopoiesis model proposed with the advent of scRNA-seq.
Until recently, multi-omic techniques linking transcriptomics with proteomics were primarily limited to surface protein analysis. However, emerging techniques such as single-cell protein and RNA co-profiling (SPARC)62 are now enabling the analysis of intracellular proteins as well.
Integrating mitochondrial genomics with transcriptomics or epigenomics
Clonal relationships can be inferred by enriching mitochondrial RNA that is already captured in low amounts by standard scRNA-seq platforms. Techniques such as mitochondrial alteration enrichment from single-cell transcriptomes to establish relatedness (MAESTER)63 have demonstrated the feasibility of this approach. Similarly, integrating scATAC-seq with mitochondrial genotyping has proven effective for inferring clonal structures. For instance, the single-cell regulatory multimaps with deep mitochondrial mutation profiling (ReDeeM)64 technique uses deep sequencing to profile mitochondrial DNA while simultaneously performing scRNA-seq and scATAC-seq, easily integrating the clonal analysis into the investigation of transcriptomic and regulatory landscapes.
Summary
The past decade has witnessed a significant expansion in the capabilities of single-cell analytics, greatly enhancing our understanding of the biology underlying hematopoiesis and hematologic malignancies. Advances in single-cell multi-omics technologies provided unprecedented insights into cellular heterogeneity, lineage differentiation, and molecular mechanisms driving disease progression. Such rapid technological progress, coupled with the decreasing cost of sequencing, is poised to further improve cellular throughput and accessibility of these technologies. Despite the current predominance of single-cell tools in research settings, there is a growing potential for their application in clinical practice. For instance, platforms such as scDNA-seq and its multi-omic adaptations could be used to identify minimal residual disease, monitor clonal evolution during therapy, and tailor personalized treatment strategies in leukemia patients.19,65 However, translating these tools to clinical use will require rigorous clinical validation and evidence that they enhance clinical decision-making.
The remarkable advancements observed over the past decade suggest that integrating single-cell technologies into clinical workflows is not far off. Continued investment and research in single-cell analytics are essential to making these tools accessible and routine in both research and clinical settings.
Footnotes
- Received August 5, 2024
- Accepted January 29, 2025
Correspondence
Disclosures
KT has received consulting fees from Celgene, GSK and Novartis, is on the scientific advisory board for Symbio Pharmaceuticals and has received honoraria from Mission Bio, Illumina, Otsuka Pharmaceuticals and Pfizer.
Contributions
YJK and KT wrote the manuscript and approved the final version.
Funding
This study was supported by National Institutes of Health grants R01CA237291 (KT) and P01CA265748 (KT), a Leukemia and Lymphoma Society Scholar Award (KT), the Breakthrough Cancer Foundation (KT), and an MD Anderson Cancer Center support grant (NIH/NCI P30 CA016672).
References
- Liggett LA, Sankaran VG. Unraveling hematopoiesis through the lens of genomics. Cell. 2020; 182(6):1384-1400. Google Scholar
- Pei S, Pollyea DA, Gustafson A. Monocytic subclones confer resistance to venetoclax-based therapy in patients with acute myeloid leukemia. Cancer Discov. 2020; 10(4):536-551. Google Scholar
- Tang F, Barbacioru C, Wang Y. mRNA-seq whole-transcriptome analysis of a single cell. Nat Methods. 2009; 6(5):377-382. Google Scholar
- Islam S, Kjällquist U, Moliner A. Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq. Genome Res. 2011; 21(7):1160-1167. Google Scholar
- Navin N, Kendall J, Troge J. Tumour evolution inferred by single-cell sequencing. Nature. 2011; 472(7341):90-94. Google Scholar
- Pollen AA, Nowakowski TJ, Shuga J. Low-coverage singlecell mRNA sequencing reveals cellular heterogeneity and activated signaling pathways in developing cerebral cortex. Nat Biotechnol. 2014; 32(10):1053-1058. Google Scholar
- Macosko EZ, Basu A, Satija R. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 2015; 161(5):1202-1214. Google Scholar
- Klein AM, Mazutis L, Akartuna I. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell. 2015; 161(5):1187-1201. Google Scholar
- Zheng GXY, Terry JM, Belgrader P. Massively parallel digital transcriptional profiling of single cells. Nat Commun. 2017; 8(1):14049. Google Scholar
- Cusanovich DA, Daza R, Adey A. Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing. Science. 2015; 348(6237):910-914. Google Scholar
- Gierahn TM, Wadsworth MH, Hughes TK. Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput. Nat Methods. 2017; 14(4):395-398. Google Scholar
- Ding J, Adiconis X, Simmons SK. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat Biotechnol. 2020; 38(6):737-746. Google Scholar
- Islam S, Zeisel A, Joost S. Quantitative single-cell RNA-seq with unique molecular identifiers. Nat Methods. 2014; 11(2):163-166. Google Scholar
- Stoeckius M, Zheng S, Houck-Loomis B. Cell hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 2018; 19(1):224. Google Scholar
- Kang HM, Subramaniam M, Targ S. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat Biotechnol. 2018; 36(1):89-94. Google Scholar
- Picelli S, Björklund ÅK, Faridani OR, Sagasser S, Winberg G, Sandberg R. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat Methods. 2013; 10(11):1096-1098. Google Scholar
- Hagemann-Jensen M, Ziegenhain C, Chen P. Single-cell RNA counting at allele and isoform resolution using Smart-seq3. Nat Biotechnol. 2020; 38(6):708-714. Google Scholar
- Byrne A, Beaudin AE, Olsen HE. Nanopore long-read RNAseq reveals widespread transcriptional variation among the surface receptors of individual B cells. Nat Commun. 2017; 8(1):16027. Google Scholar
- Takahashi K, Tanaka T. Clonal evolution and hierarchy in myeloid malignancies. Trends Cancer. 2023; 9(9):707-715. Google Scholar
- Telenius H, Carter NP, Bebb CE, Nordenskjöld M, Ponder BA, Tunnacliffe A. Degenerate oligonucleotide-primed PCR: general amplification of target DNA by a single degenerate primer. Genomics. 1992; 13(3):718-725. Google Scholar
- Zong C, Lu S, Chapman AR, Xie XS. Genome-wide detection of single-nucleotide and copy-number variations of a single human cell. Science. 2012; 338(6114):1622-1626. Google Scholar
- Dean FB, Hosono S, Fang L. Comprehensive human genome amplification using multiple displacement amplification. Proc Natl Acad Sci U S A. 2002; 99(8):5261-5266. Google Scholar
- Gonzalez-Pena V, Natarajan S, Xia Y. Accurate genomic variant detection in single cells with primary template-directed amplification. Proc Natl Acad Sci U S A. 2021; 118(24):e2024176118. Google Scholar
- Hashimshony T, Wagner F, Sher N, Yanai I. CEL-seq: single-cell RNA-seq by multiplexed linear amplification. Cell Rep. 2012; 2(3):666-673. Google Scholar
- Hou Y, Song L, Zhu P. Single-Cell Exome Single-cell exome sequencing and monoclonal evolution of a JAK2-negative myeloproliferative neoplasm. Cell. 2012; 148(5):873-885. Google Scholar
- Xu X, Hou Y, Yin X. Single-cell exome sequencing reveals single-nucleotide mutation characteristics of a kidney tumor. Cell. 2012; 148(5):886-895. Google Scholar
- Lee-Six H, Øbro NF, Shepherd MS. Population dynamics of normal human blood inferred from somatic mutations. Nature. 2018; 561(7724):473-478. Google Scholar
- Mitchell E, Spencer Chapman M, Williams N. Clonal dynamics of haematopoiesis across the human lifespan. Nature. 2022; 606(7913):343-350. Google Scholar
- Fabre MA, de Almeida JG, Fiorillo E. The longitudinal dynamics and natural history of clonal haematopoiesis. Nature. 2022; 606(7913):335-342. Google Scholar
- Williams N, Lee J, Mitchell E. Life histories of myeloproliferative neoplasms inferred from phylogenies. Nature. 2022; 602(7895):162-168. Google Scholar
- Wang Y, Waters J, Leung ML. Clonal evolution in breast cancer revealed by single nucleus genome sequencing. Nature. 2014; 512(7513):155-160. Google Scholar
- Demaree B, Delley CL, Vasudevan HN. Joint profiling of DNA and proteins in single cells to dissect genotype-phenotype associations in leukemia. Nat Commun. 2021; 12(1):1583. Google Scholar
- Pellegrino M, Sciambi A, Treusch S. High-throughput single-cell DNA sequencing of acute myeloid leukemia tumors with droplet microfluidics. Genome Res. 2018; 28(9):1345-1352. Google Scholar
- Marks JR, Zawistowski JS, Salas-González I. Unifying comprehensive genomics and transcriptomics in individual cells to illuminate oncogenic and drug resistance mechanisms. bioRxiv. 2023. Google Scholar
- Ludwig LS, Lareau CA, Ulirsch JC. Lineage tracing in humans enabled by mitochondrial mutations and single-cell genomics. Cell. 2019; 176(6):1325-1339.e1322. Google Scholar
- Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods. 2013; 10(12):1213-1218. Google Scholar
- You Y, Tian L, Su S. Benchmarking UMI-based single-cell RNA-seq preprocessing workflows. Genome Biol. 2021; 22(1):339. Google Scholar
- Heumos L, Schaar AC, Lance C. Best practices for singlecell analysis across modalities. Nat Rev Genet. 2023; 24(8):550-572. Google Scholar
- Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015; 33(5):495-502. Google Scholar
- Wolf FA, Angerer P, Theis FJ. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 2018; 19(1):15. Google Scholar
- Rodriguez-Meira A, Buck G, Clark SA. Unravelling intratumoral heterogeneity through high-sensitivity single-cell mutational analysis and parallel RNA sequencing. Mol Cell. 2019; 73(6):1292-1305.e8. Google Scholar
- Jakobsen NA, Turkalj S, Zeng AGX. Selective advantage of mutant stem cells in human clonal hematopoiesis is associated with attenuated response to inflammation and aging. Cell Stem Cell. 2024; 31(8):1127-1144.e17. Google Scholar
- Patel AP, Tirosh I, Trombetta JJ. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014; 344(6190):1396-1401. Google Scholar
- Gao R, Bai S, Henderson YC. Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes. Nat Biotechnol. 2021; 39(5):599-608. Google Scholar
- van Galen P, Hovestadt V, Wadsworth Ii MH. Single-cell RNA-seq reveals AML hierarchies relevant to disease progression and immunity. Cell. 2019; 176(6):1265-1281.e24. Google Scholar
- Nam AS, Kim K-T, Chaligne R. Somatic mutations and cell identity linked by genotyping of transcriptomes. Nature. 2019; 571(7765):355-360. Google Scholar
- Thijssen R, Tian L, Anderson MA. Single-cell multiomics reveal the scale of multilayered adaptations enabling CLL relapse during venetoclax therapy. Blood. 2022; 140(20):2127-2141. Google Scholar
- Guo H, Zhu P, Wu X, Li X, Wen L, Tang F. Single-cell methylome landscapes of mouse embryonic stem cells and early embryos analyzed using reduced representation bisulfite sequencing. Genome Res. 2013; 23(12):2126-2135. Google Scholar
- Turkalj S, Jakobsen NA, Groom A. GTAC enables parallel genotyping of multiple genomic loci with chromatin accessibility profiling in single cells. Cell Stem Cell. 2023; 30(5):722-740.e11. Google Scholar
- Izzo F, Myers RM, Ganesan S. Mapping genotypes to chromatin accessibility profiles in single cells. Nature. 2024; 629(8014):1149-1157. Google Scholar
- Mimitou EP, Lareau CA, Chen KY. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat Biotechnol. 2021; 39(10):1246-1258. Google Scholar
- Hou Y, Guo H, Cao C. Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res. 2016; 26(3):304-319. Google Scholar
- Charlton J, Downing TL, Smith ZD. Global delay in nascent strand DNA methylation. Nat Struct Mol Biol. 2018; 25(4):327-332. Google Scholar
- Gaiti F, Chaligne R, Gu H. Epigenetic evolution and lineage histories of chronic lymphocytic leukaemia. Nature. 2019; 569(7757):576-580. Google Scholar
- Shahi P, Kim SC, Haliburton JR, Gartner ZJ, Abate AR. Abseq: ultrahigh-throughput single cell protein profiling with droplet microfluidic barcoding. Sci Rep. 2017; 7(1):44447. Google Scholar
- Zhang X, Song B, Carlino MJ. An immunophenotype-coupled transcriptomic atlas of human hematopoietic progenitors. Nat Immunol. 2024; 25(4):703-715. Google Scholar
- Morita K, Wang F, Jahn K. Clonal evolution of acute myeloid leukemia revealed by high-throughput single-cell genomics. Nat Commun. 2020; 11(1):5327. Google Scholar
- Miles LA, Bowman RL, Merlinsky TR. Single-cell mutation analysis of clonal evolution in myeloid malignancies. Nature. 2020; 587(7834):477-482. Google Scholar
- Lachowiez CA, Loghavi S, Zeng Z. A phase Ib/II study of ivosidenib with venetoclax ± azacitidine in IDH1-mutated myeloid malignancies. Blood Cancer Discov. 2023; 4(4):276-293. Google Scholar
- Dillon LW, Ghannam J, Nosiri C. Personalized single-cell proteogenomics to distinguish acute myeloid leukemia from nonmalignant clonal hematopoiesis. Blood Cancer Discov. 2021; 2(4):319-325. Google Scholar
- Stoeckius M, Hafemeister C, Stephenson W. Simultaneous epitope and transcriptome measurement in single cells. Nat Methods. 2017; 14(9):865-868. Google Scholar
- Reimegård J, Tarbier M, Danielsson M. A combined approach for single-cell mRNA and intracellular protein expression analysis. Commun Biol. 2021; 4(1):624. Google Scholar
- Miller TE, Lareau CA, Verga JA. Mitochondrial variant enrichment from high-throughput single-cell RNA sequencing resolves clonal populations. Nat Biotechnol. 2022; 40(7):1030-1034. Google Scholar
- Weng C, Yu F, Yang D. Deciphering cell states and genealogies of human haematopoiesis. Nature. 2024; 627(8003):389-398. Google Scholar
- Robinson TM, Bowman RL, Persaud S. Single-cell genotypic and phenotypic analysis of measurable residual disease in acute myeloid leukemia. Sci Adv. 2023; 9(38):eadg0488. Google Scholar
Figures & Tables
Article Information

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.