Abstract

As the demographic structure shifts towards an aging society, strategies aimed at slowing down or reversing the aging process become increasingly essential. Aging is a major predisposing factor for many chronic diseases in humans. The hematopoietic system, comprising blood cells and their associated bone marrow microenvironment, intricately participates in hematopoiesis, coagulation, immune regulation and other physiological phenomena. The aging process triggers various alterations within the hematopoietic system, serving as a spectrum of risk factors for hematopoietic disorders, including clonal hematopoiesis, immune senescence, myeloproliferative neoplasms and leukemia. The emerging single-cell technologies provide novel insights into age-related changes in the hematopoietic system. In this review, we summarize recent studies dissecting hematopoietic system aging using single-cell technologies. We discuss cellular changes occurring during aging in the hematopoietic system at the levels of the genomics, transcriptomics, epigenomics, proteomics, metabolomics and spatial multi-omics. Finally, we contemplate the future prospects of single-cell technologies, emphasizing the impact they may bring to the field of hematopoietic system aging research.

INTRODUCTION

Aging constitutes a major susceptibility factor for numerous chronic maladies in humans, including hematologic disorders, cardiovascular ailments, cancer and neurodegenerative conditions. The aging process entails substantial alterations in cellular constitution, organ dimensions and functionality across diverse tissues and organs within an organism. Notably, the hematopoietic system, comprising blood cells and the associated bone marrow niche, intricately participates in hematopoiesis, coagulation, immune modulation and other physiological phenomena [1, 2]. Hematopoietic stem cells (HSCs) exhibit the remarkable capacity for both self-renewal and differentiation, ensuring the perpetual production of the entire spectrum of blood cells throughout their life cycle. The aging process instigates manifold transformations within the hematopoietic system. Extant research delineates that age-associated changes in the bone marrow microenvironment and HSCs engender disruptions in tissue homeostasis, manifesting in discernible disparities between youthful and aged cohorts. These disparities encompass shifts in total cellular quantity, lineage-specific differentiations, cellular constitution and functional attributes of HSCs [3–6]. These are a series of risk factors leading to hematopoietic diseases, including clonal hematopoiesis, immune aging, myeloproliferative neoplasms and leukemia [7–9]. Consequently, unraveling the intricate molecular mechanisms underlying the aging of HSCs assumes paramount importance. This endeavor holds the key to identifying potential intervention targets aimed at prolonging hematopoietic healthspan. Moreover, it bears relevance in the therapeutic landscape for addressing immune system deficiencies, age-related inflammatory pathologies and hematologic maladies.

In the realm of experimental interpretations, both in vitro and in vivo, a prevailing assumption persist, namely the uniformity of utilized cells or tissues. However, such a conventional approach often neglects inherent cellular variances, resulting in the inadvertent loss of crucial information. The complex multitude of various cell types in hematopoietic system, from HSCs to mature cells, within both normal physiological processes and pathological states, makes the research on it particularly challenging. In the preceding century, pioneers in the scientific community initiated the sequencing of the entire transcriptome at the single-cell level [10]. In 2009, Tang laid the foundation for the single-cell mRNA-Seq method [11]. Over the ensuing decade, as this technology matured and became commercially accessible, it underwent rapid evolution. What was once confined to specialized research laboratories has now proliferated into a diverse array of commercial applications. While single-cell transcriptomics stands as an exemplary tool for quantifying gene expression variations at the single-cell tier, it has become increasingly evident that it falls short of fully addressing the diverse requirements of researchers across multifarious domains. Over this decade-long trajectory, single-cell technology has not merely transcended the bounds of transcriptomics; it has extended its purview into genomics, metabolomics, epigenomics and beyond [12–15]. Single-cell technology has been used in many fields, such as analyzing steady-state cardiovascular cell diversity and cardiovascular disease targets [16], explaining the molecular mechanisms and spatial characteristics of brain aging [17, 18] and revealing the mechanisms of cancer [19–22].

Single-cell technology, with its unparalleled capacity to unravel intricate cellular compositions, serves as a potent instrument for dissecting the aging dynamics of the hematopoietic system, thus advancing our comprehension of the underlying mechanisms of aging. This review is focalized on recent revelations made possible by single-cell technologies in the investigation of aging dynamics spanning diverse lineages within the hematopoietic system across multiple species. We present a brief overview of each single-cell technique and its applications in studying hematopoietic aging (Table 1) and a timeline highlighting key findings from single-cell studies of the hematopoietic system (Fig. 1).

Table 1

Overview of single-cell technology and its applications in hematopoietic aging studies

MethodMeasurementYearAdvantageApplication in hematopoietic system aging
scDNAseqgenome2011Provides genome-wide information including CNVs and SNVs at the single-cell levelUnravelling Intratumoral Heterogeneity [23].
Linking somatic mutations to cell identity [24].
Analysis of Intratumoral Heterogeneity in MDS with Isolated del(5q) [25].
Unveiling LCP1 and WNK1 mutations as drivers of CLL [26].
MtDNA mutations may affect mitochondrial function [27].
scRNAseqtranscriptome2011Provides gene expression information in single cells and solves the problem of cell heterogeneity existing in traditional methodsChanges in cell cycle and differentiation programs [28, 29].
Aging correlates with an upregulation of platelet-specific genes [30].
Reveal a concomitant delay in differentiation and cell cycle of aged HSCs [31].
Characteristics of aging in HSCs [32].
Epigenetically altered genes are downregulated during age [33].
SCtranscriptome to Characterize the Therapeutic Response of CML [34].
The lineage bias of HSPCs was correlated with glycolysis-related pathways [35].
scATAC-seqchromosome conformation2015This technology uses Tn5 transposase to cleave regions of DNA that are not protected by binding proteins and is used to analyze chromatin openness and measure dynamic changes in chromatin structure.DNA methylase plays a key role in maintaining HSC homeostasis [36].
Age-related changes in chromatin configuration influence the mode of HSC division. [37]
scBS-Seqhistone modification2020A method for single-cell DNA methylation analysis at single-base resolutionRevealing the presence of distinct epigenetic subpopulations [38].
TARGET-seqgenome
transcriptome
2020focuses on improving coverage of key mutations.Revealing cells outside the MPN clone also exhibit aberrant gene expression [39].
CITE-seqtranscriptome
proteome
2017Provides detection of cell surface protein information and intracellular transcriptome information at the single cell level.An immunophenotype-coupled transcriptomic atlas of human hematopoietic progenitors [40].
scHi-Cchromosome conformation2013Provides information on the 3D structure of the entire genome in a single cell.
DR-seqgenome
transcriptome
2015Concurrently amplifies DNA and RNA, allowing for RNA-seq and DNA-seq applications. It is a plate-based method with low throughput, designed to mitigate the risk of nucleic acid loss.
G&T-seqgenome
transcriptome
2015Concurrently analyzes DNA and RNA and can sequence the full length of RNA.
MethodMeasurementYearAdvantageApplication in hematopoietic system aging
scDNAseqgenome2011Provides genome-wide information including CNVs and SNVs at the single-cell levelUnravelling Intratumoral Heterogeneity [23].
Linking somatic mutations to cell identity [24].
Analysis of Intratumoral Heterogeneity in MDS with Isolated del(5q) [25].
Unveiling LCP1 and WNK1 mutations as drivers of CLL [26].
MtDNA mutations may affect mitochondrial function [27].
scRNAseqtranscriptome2011Provides gene expression information in single cells and solves the problem of cell heterogeneity existing in traditional methodsChanges in cell cycle and differentiation programs [28, 29].
Aging correlates with an upregulation of platelet-specific genes [30].
Reveal a concomitant delay in differentiation and cell cycle of aged HSCs [31].
Characteristics of aging in HSCs [32].
Epigenetically altered genes are downregulated during age [33].
SCtranscriptome to Characterize the Therapeutic Response of CML [34].
The lineage bias of HSPCs was correlated with glycolysis-related pathways [35].
scATAC-seqchromosome conformation2015This technology uses Tn5 transposase to cleave regions of DNA that are not protected by binding proteins and is used to analyze chromatin openness and measure dynamic changes in chromatin structure.DNA methylase plays a key role in maintaining HSC homeostasis [36].
Age-related changes in chromatin configuration influence the mode of HSC division. [37]
scBS-Seqhistone modification2020A method for single-cell DNA methylation analysis at single-base resolutionRevealing the presence of distinct epigenetic subpopulations [38].
TARGET-seqgenome
transcriptome
2020focuses on improving coverage of key mutations.Revealing cells outside the MPN clone also exhibit aberrant gene expression [39].
CITE-seqtranscriptome
proteome
2017Provides detection of cell surface protein information and intracellular transcriptome information at the single cell level.An immunophenotype-coupled transcriptomic atlas of human hematopoietic progenitors [40].
scHi-Cchromosome conformation2013Provides information on the 3D structure of the entire genome in a single cell.
DR-seqgenome
transcriptome
2015Concurrently amplifies DNA and RNA, allowing for RNA-seq and DNA-seq applications. It is a plate-based method with low throughput, designed to mitigate the risk of nucleic acid loss.
G&T-seqgenome
transcriptome
2015Concurrently analyzes DNA and RNA and can sequence the full length of RNA.

CNVs, copy number variations; SNVs, single nucleotide variations; HSC, hematopoietic stem cell; mtDNA, mitochondrial DNA; MDS, myelodysplastic syndromes; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia; MPN, myeloproliferative neoplasms; HSPC, hematopoietic stem and progenitor cells; scDNAseq, single-cell DNA sequencing; scRNAseq, single-cell RNA sequencing; scATAC-seq, single-cell assay for transposase accessible chromatin sequencing; scBS-seq, single-cell bisulphite sequencing; CITE-seq, cellular indexing of transcriptomes and epitopes by sequencing; scHi-C, Single-cell high-throughput chromosome conformation capture; DR-seq, gDNA-mRNA sequencing; G&T-seq, genome and transcriptome sequencing.

Table 1

Overview of single-cell technology and its applications in hematopoietic aging studies

MethodMeasurementYearAdvantageApplication in hematopoietic system aging
scDNAseqgenome2011Provides genome-wide information including CNVs and SNVs at the single-cell levelUnravelling Intratumoral Heterogeneity [23].
Linking somatic mutations to cell identity [24].
Analysis of Intratumoral Heterogeneity in MDS with Isolated del(5q) [25].
Unveiling LCP1 and WNK1 mutations as drivers of CLL [26].
MtDNA mutations may affect mitochondrial function [27].
scRNAseqtranscriptome2011Provides gene expression information in single cells and solves the problem of cell heterogeneity existing in traditional methodsChanges in cell cycle and differentiation programs [28, 29].
Aging correlates with an upregulation of platelet-specific genes [30].
Reveal a concomitant delay in differentiation and cell cycle of aged HSCs [31].
Characteristics of aging in HSCs [32].
Epigenetically altered genes are downregulated during age [33].
SCtranscriptome to Characterize the Therapeutic Response of CML [34].
The lineage bias of HSPCs was correlated with glycolysis-related pathways [35].
scATAC-seqchromosome conformation2015This technology uses Tn5 transposase to cleave regions of DNA that are not protected by binding proteins and is used to analyze chromatin openness and measure dynamic changes in chromatin structure.DNA methylase plays a key role in maintaining HSC homeostasis [36].
Age-related changes in chromatin configuration influence the mode of HSC division. [37]
scBS-Seqhistone modification2020A method for single-cell DNA methylation analysis at single-base resolutionRevealing the presence of distinct epigenetic subpopulations [38].
TARGET-seqgenome
transcriptome
2020focuses on improving coverage of key mutations.Revealing cells outside the MPN clone also exhibit aberrant gene expression [39].
CITE-seqtranscriptome
proteome
2017Provides detection of cell surface protein information and intracellular transcriptome information at the single cell level.An immunophenotype-coupled transcriptomic atlas of human hematopoietic progenitors [40].
scHi-Cchromosome conformation2013Provides information on the 3D structure of the entire genome in a single cell.
DR-seqgenome
transcriptome
2015Concurrently amplifies DNA and RNA, allowing for RNA-seq and DNA-seq applications. It is a plate-based method with low throughput, designed to mitigate the risk of nucleic acid loss.
G&T-seqgenome
transcriptome
2015Concurrently analyzes DNA and RNA and can sequence the full length of RNA.
MethodMeasurementYearAdvantageApplication in hematopoietic system aging
scDNAseqgenome2011Provides genome-wide information including CNVs and SNVs at the single-cell levelUnravelling Intratumoral Heterogeneity [23].
Linking somatic mutations to cell identity [24].
Analysis of Intratumoral Heterogeneity in MDS with Isolated del(5q) [25].
Unveiling LCP1 and WNK1 mutations as drivers of CLL [26].
MtDNA mutations may affect mitochondrial function [27].
scRNAseqtranscriptome2011Provides gene expression information in single cells and solves the problem of cell heterogeneity existing in traditional methodsChanges in cell cycle and differentiation programs [28, 29].
Aging correlates with an upregulation of platelet-specific genes [30].
Reveal a concomitant delay in differentiation and cell cycle of aged HSCs [31].
Characteristics of aging in HSCs [32].
Epigenetically altered genes are downregulated during age [33].
SCtranscriptome to Characterize the Therapeutic Response of CML [34].
The lineage bias of HSPCs was correlated with glycolysis-related pathways [35].
scATAC-seqchromosome conformation2015This technology uses Tn5 transposase to cleave regions of DNA that are not protected by binding proteins and is used to analyze chromatin openness and measure dynamic changes in chromatin structure.DNA methylase plays a key role in maintaining HSC homeostasis [36].
Age-related changes in chromatin configuration influence the mode of HSC division. [37]
scBS-Seqhistone modification2020A method for single-cell DNA methylation analysis at single-base resolutionRevealing the presence of distinct epigenetic subpopulations [38].
TARGET-seqgenome
transcriptome
2020focuses on improving coverage of key mutations.Revealing cells outside the MPN clone also exhibit aberrant gene expression [39].
CITE-seqtranscriptome
proteome
2017Provides detection of cell surface protein information and intracellular transcriptome information at the single cell level.An immunophenotype-coupled transcriptomic atlas of human hematopoietic progenitors [40].
scHi-Cchromosome conformation2013Provides information on the 3D structure of the entire genome in a single cell.
DR-seqgenome
transcriptome
2015Concurrently amplifies DNA and RNA, allowing for RNA-seq and DNA-seq applications. It is a plate-based method with low throughput, designed to mitigate the risk of nucleic acid loss.
G&T-seqgenome
transcriptome
2015Concurrently analyzes DNA and RNA and can sequence the full length of RNA.

CNVs, copy number variations; SNVs, single nucleotide variations; HSC, hematopoietic stem cell; mtDNA, mitochondrial DNA; MDS, myelodysplastic syndromes; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia; MPN, myeloproliferative neoplasms; HSPC, hematopoietic stem and progenitor cells; scDNAseq, single-cell DNA sequencing; scRNAseq, single-cell RNA sequencing; scATAC-seq, single-cell assay for transposase accessible chromatin sequencing; scBS-seq, single-cell bisulphite sequencing; CITE-seq, cellular indexing of transcriptomes and epitopes by sequencing; scHi-C, Single-cell high-throughput chromosome conformation capture; DR-seq, gDNA-mRNA sequencing; G&T-seq, genome and transcriptome sequencing.

Selected examples of research achievements on hematopoietic system aging assisted by single-cell omics technology. The panels above and below the timeline showcase significant findings obtained over the last years in the exploration of hematopoietic system aging through single-cell sequencing technology.
Figure 1

Selected examples of research achievements on hematopoietic system aging assisted by single-cell omics technology. The panels above and below the timeline showcase significant findings obtained over the last years in the exploration of hematopoietic system aging through single-cell sequencing technology.

GENOMIC ALTERATIONS CORRELATED WITH HEMATOPOIETIC AGING

The accrual of DNA damage is commonly acknowledged as a characteristic feature of the aging process [41]. With advancing age, there is a gradual accrual of somatic mutations across tissues. While the majority of these mutations may have minimal functional impact, some mutations can confer new phenotypes on cells, such as an increased rate of division. In the hematopoietic system, specific functional changes can occur when certain HSCs acquire mutations that enhance their proliferation ability, leading to the development of a substantial number of mature blood cells—a phenomenon known as clonal hematopoiesis [42, 43]. Research indicates that clonal hematopoiesis is associated with aging: Somatic clones are detectable in a small proportion of healthy individuals under the age of 40, but the likelihood of their occurrence rises with age [44]. Clonal hematopoiesis is also linked to an elevated risk of hematological malignancies [45, 46]. Hematologic malignancies exhibit cellular heterogeneity, with rare cell populations, including drug-resistant cell clones and leukemia stem cells, playing a crucial role in disease progression. Given this complexity, single-cell genomics approaches are particularly suitable for studying these malignancies [47].

Genomic DNA

Single-cell techniques are currently employed to investigate the association between somatic mutations and age-related diseases. Unlike single-cell RNA sequencing (scRNA-seq), the advancement of single-cell DNA sequencing (scDNA-seq) methods poses a greater challenge due to the inherent complexity of dealing with only two copies of genomic DNA in a single cell. In 2012, Zong [48] pioneered the development of multiple unified whole-genome amplification (WGA) methods, including multiple annealing and loop-based amplification cycles (MALBAC), enabling single-cell whole-genome sequencing (scWGS). Moreover, several methods have emerged to simultaneously perform scRNA-seq and scDNA-seq on individual cells. Instances of such methods include DR-Seq and G&T-seq, both relying on comprehensive cell lysis followed by genome isolation [49, 50]. Additionally, techniques such as isolation of polyadenylated RNA from DNA and direct nuclear labeling and RNA-seq have been employed in this context [51, 52].

TARGET-seq facilitates simultaneous mutation and RNA sequencing analysis in single cells. It uses flow cytometry combined with cell surface proteomics to correlate gene expression and mutational status within specialized cell populations [39]. An examination of non-mutated cellular HSCs has yielded valuable insights into the disruption of normal hematopoiesis in myeloproliferative neoplasms (MPNs), revealing that cells outside the MPN clone also exhibit aberrant gene expression [23]. Genotyping of Transcriptomes (GoT) is presently employed to distinguish mutation-positive cells from mutation-negative cells of the same type. The simultaneous analysis of mutation status and transcriptome using GoT was initially applied to hematopoietic stem and progenitor cells (HSPCs) from CALR-mutated essential thrombocythemia (ET) patients [24]. A study unveils intertumor heterogeneity by conducting single-cell copy number analysis and genotyping of HSPCs isolated from the bone marrow of patients with myelodysplastic syndromes (MDS) [25]. Another study employed targeted single-cell sequencing of whole myeloid cells and multipotent progenitor cells to unveil the genetic foundation of disease initiation in T-cell acute lymphoblastic leukemia (T-ALL), and found that mutation in T-ALL may originate from multipotent progenitor cells [53]. In another study, single-cell genome sequencing was employed to delineate clonal evolution in T-ALL patients during diagnosis and treatment, and revealed that minor clones evolved into clinically relevant major clones later in the disease course [54]. Utilizing single-cell genome and transcriptome analyses, a study proposes that mutations in LCP1 and WNK1 could represent novel mechanisms propelling the development of chronic lymphocytic leukemia (CLL) [26]. Gawad sequenced individual tumor cells from patients with ALL and detected co-dominant clones in the majority of cases. This sequencing approach offers insights into the disease at a single-cell resolution, paving the way for potential novel treatment strategies [55].

Mitochondrial DNA

Each cell harbors hundreds to thousands of mitochondrial DNA (mtDNA) copies, which replicate independently of nuclear genomic DNA [56]. In comparison to nuclear DNA, mtDNA accumulates mutations at a rate 5–10 times higher, primarily attributed to the inability to undergo DNA repair and frequent exposure to reactive oxygen species [57], contrary to the conventional belief that individuals predominantly possess identical mitochondrial genomes. Currently, research is underway to explore the origins of human mtDNA variations and their association with human diseases, encompassing cancer, neurodegenerative diseases and aging [58–62]. Notably, hundreds of mtDNA mutations have been identified as causes of various human diseases [63]. However, the contribution of mtDNA mutations to these processes remains unclear, and studying mtDNA mutations and their associated pathogenic effects at the single-cell level is essential [64]. Several methods have been developed to detect mtDNA mutations at the single-cell level [27, 65–69]. One study performed a systematic evaluation of mtDNA mutations in B lymphocytes and monocytes of an elderly individual at the single-cell level, and found that over 50% of cells carried mtDNA mutations, and over 30% those mutations were predicted to be highly pathogenic, suggesting that mtDNA mutations may constitute a significant source of mitochondrial function decline with age [27]. Studies demonstrated higher mtDNA biosynthesis in acute myeloid leukemia (AML) blast cells compared to normal hematopoietic cells [70]. Resistance to imatinib in chronic myelogenous leukemia (CML) is linked to heightened instability in mtDNA and elevated levels of reactive oxygen species [71]. The integrity of mitochondria is crucial to HSC function, and mutations in mtDNA may lead to diseases. While clear mitochondrial abnormalities are observed in leukemia cells, their role in leukemia development and progression remains relatively unknown. Single-cell sequencing analysis enables the tracking of specific HSC clones and reveals mutation patterns in hematopoietic cells at the single-cell level, which will provide new insights into the occurrence of mtDNA mutations in HSCs and enhance our understanding of their role in aging and disease.

TRANSCRIPTOMIC ALTERATIONS CORRELATED WITH HEMATOPOIETIC AGING

Comparing the transcriptome of cells with disparate ages within the same cell type is a common strategy to investigate the molecular mechanisms underlying aging. Given the hematopoietic system’s diversity in cell populations, leveraging single-cell transcriptomics provides a necessary and valuable approach. Single-cell level analysis can determine whether age-related changes occur in specific cell type or homogeneously present across all cell populations.

Age-related gene expression changes in HSCs

Before the widespread adoption of single-cell sequencing technology, considerable attention was devoted to investigating alterations in HSC function during aging, yet the underlying molecular mechanisms remained incompletely understood. Presently, numerous studies leverage single-cell sequencing to scrutinize the transcriptome of HSCs across various age groups, providing more precise insights. This approach aids in unraveling age-specific changes in HSC gene expression, exposing factors like lineage bias, imbalances in self-renewal and differentiation, and the molecular mechanisms underpinning phenotypes.

Transcriptomic analysis at the single-cell level offers distinct advantages in elucidating the phenotypic changes in HSCs during aging. Researchers frequently employ single-cell techniques to investigate HSC function in mouse models. Single-cell RNA-seq (scRNA-seq) was used to analyze transcriptome changes in HSPCs in young and old mice. The findings revealed that the variability within each cell population was predominantly influenced by the cell cycle. Notably, the number of cells in the G1 phase was lower in old long-term hematopoietic stem cells (LTHSCs) compared to young LTHSCs, suggesting an accelerated progression through the G1 phase, which is consistent with models proposing that changes in cell cycle progression in relation to HSC self-renewal and differentiation are associated with aging [28, 29]. In another study, a thorough analysis of the single-cell transcriptomes and function of mouse HSCs revealed that aging correlates with an upregulation of platelet-specific genes in HSCs. [30]. Hérault conducted a comparative analysis of HSPCs in young and old mice, revealing alterations in the expression of genes associated with loss of differentiation and heightened hemostatic characteristics during aging. Notable changes included the upregulation of Nupr1, Vwf and Clu and undifferentiated HSPC markers, along with the downregulation of HSC differentiation genes. These findings align with previously reported shifts in the differentiation potential and platelet bias of aged HSPCs [31]. One study compiled 16 published and unpublished datasets, determining that dysregulated senescence genes predominantly consist of membrane-associated transcripts, including numerous cell surface molecules not previously linked to HSC biology. Selp is not only associated with reduced HSC function but also serves as a marker of aging HSCs. Furthermore, single-cell transcriptome analysis revealed the presence of a specialized population of HSCs in aging bone marrow with transcriptional profiles similar to those of young HSCs [72]. In a previous study, Kirschner characterized the heterogeneous aging of HSCs through single-cell transcriptome sequencing analysis, identifying a subset of aged HSCs exhibiting signs of functional failure. The expansion of the aged HSC subpopulation through increased proliferation was linked to decreased HSC function, underscoring the connection between prolonged proliferation and diminished HSC functionality [73]. Studying gene expression at the isoform level is required to fully understand transcriptional cellular heterogeneity. ScRNA-seq can annotate cells and cell types with isotype-level expression information, allowing a comprehensive understanding of the complexity of cellular transcriptional and functional phenotypes. In a recent study, Mincarelli et al. presented an integrated short- and long-read single-cell RNA-seq analysis of haematopoietic stem and progenitor cells. They demonstrated that over half of detected genes are expressed as multiple, often functionally distinct, isoforms, including many transcription factors and key cytokine receptors. Such an approach not only facilitates the integration of previously unlabeled isoforms into reference transcriptomes but also enhances the accuracy of annotating cell type-specific isoform expression [32].

In addition to single-cell analysis in mouse models, researchers also directed their attention to human bone marrow cells. The data revealed variations among healthy donors and demonstrated age-related changes in cell population frequencies [74]. ScRNA-seq is very conducive to the analysis of HSC heterogeneity, but provide limited temporal information. HSPC biology is highly dynamic, which urgently necessitates the high-resolution analysis of the quantity, differentiation, proliferation and other dynamic processes of HSCs at different time points in vivo. In a recent study, a real-time and quantitative murine bone marrow hematopoietic cell flow model was constructed based on time-series scRNA-seq and continuous labeling techniques. This model was employed to reveal various self-renewal processes and differentiation characteristics within different lineages, bridging molecular processes with cellular behavior [75]. The innovative aspect of this model lies in its connection of high-resolution molecular information with tissue-scale cellular behavior, filling a void in the lack of in vivo dynamic models in the hematopoietic system and holding potential applications in biological fields beyond hematopoiesis.

Gene expression changes in mature blood cells and age-related diseases

Age-related changes in the mature transcriptome can potentially elucidate the functional alterations observed during human aging. The Tabula Muris Consortium employed scRNA-seq to construct a comprehensive map of age-related changes in the organs and tissues of mice across different age groups. This represents one of the most extensive analyses to date of the mammalian aging process at single-cell resolution. Results showed that white blood cells from aged mice showed increased expression of pro-inflammatory markers, while decreased expression of anti-inflammatory markers [76]. Studies have also showed a 2-fold reduction in the fraction of naive CD8 T cells in peripheral blood mononuclear cells in older adults compared to younger adults [77, 78]. In a study by Hashimoto, single-cell transcriptome analysis of peripheral blood mononuclear cells from centenarians and young controls revealed that a unique subset of cytotoxic CD4 T cells was significantly increased in centenarians [79]. This subset may contribute to longevity by preserving immune responses against infection and disease. Additionally, another study reported a progressive increase in B cell clonality in aging mice [80]. The results of these single-cell studies help explore the antigenic specificity of aging-related cell subsets and their inflammatory functions in aging organisms.

The elderly population faces an elevated risk of malignant hematopoiesis [81]. Hematopoietic malignancies such as leukemia, lymphoma, multiple myeloma (MM), MPN and MDS are prevalent and pose a significant threat to patient health and survival. Employing scRNA-seq methods can contribute to a more profound understanding of these diseases and facilitate the discovery of more effective treatment strategies.

Alice and colleagues developed a method that combines highly sensitive mutation detection with whole-transcriptome analysis of the same single cells. They applied this technique to analyze single cells from CML patients, revealing the heterogeneity of CML-SCs. Furthermore, a crisis-specific stem cell population among blasts was identified, present in chronic-phase CML-SC subclones of patients who subsequently developed blast crisis [82]. Zhang conducted scRNA-seq analysis on individual cells isolated from the peripheral blood of CML patients at various stages of treatment, and found that imatinib treatment resulted in substantial alterations in leukocyte populations in both responders and non-responders, and gene expression changes in diverse cell populations, underscoring the potential utility of peripheral blood as a diagnostic tool for assessing tyrosine kinase inhibitor sensitivity in CML patients [83]. Ma integrated scRNA-seq profiles and network analysis of CML stem cells to unravel the mechanisms underlying different tyrosine kinase inhibitor responses. They found that a set of genes in various types of stem cells from CML patients was consistently differentially expressed compared to HSCs [34]. Vaidehi Krishnan identified pre-treatment signatures of primary imatinib resistance in CML through a single-cell atlas [84]. Wang discovered significant changes in multiple cellular functions of cells carrying SF3B1 mutations in CLL, affecting multiple cellular functions. They defined LCP1 and WNK1 mutations as new drivers of CLL development [85]. Van Galen analyzed AML heterogeneity using scRNA-seq, with a specific focus on FLT3 genotypes in malignant cell clusters [86]. Lohr et al. also used scRNA-seq to identify genes that are differentially expressed between circulating tumor cells from different MM patients, which can be used for the diagnosis and classification of MM [87]. By combing scRNA-seq and single-cell genotyping, Tong found that HSCs with JAK2 mutations showed a bias toward megakaryocyte differentiation, and this type of mutation was most common in MPN [88].

EPIGENETIC ALTERATIONS CORRELATED WITH AGING IN HEMATOPOIETIC SYSTEM

Epigenetics represents a reversible mechanism influencing gene expression which does not involve alterations of DNA sequence [89], and holds a pivotal role in numerous biological processes [90]. A study analyzed epigenomic and transcriptomic changes during normal human aging, and, with the help of scRNA-seq analysis, confirmed that the observed age-associated changes in the epigenetic program are unlikely to stem from the expansion of a pre-existing subpopulation within the bone marrow, but rather appear to result from true epigenetic reprogramming [33]. Epigenetic changes associated with aging encompass DNA methylation, chromatin remodeling and post-translational modifications of histones. Aging can induce alterations the epigenetic landscape within HSCs [91–93].

DNA modification

DNA methylation, the earliest studied epigenetic mark, plays a crucial role in various biological processes [94, 95]. Hui employed single-cell bisulfite sequencing (scBS-Seq) to explore the methylome of cells in the HSC compartment of both mice and humans, revealing the presence of distinct epigenetic subpopulations [38]. During the differentiation of HSCs into multipotent progenitors and cells of various lineages, DNA methylation undergoes extensive changes [96]. The aging process is associated with a widespread loss of methylation across the genome [97, 98]. In a study investigating the pathogenic mechanisms of aging in whole blood, MBD-based capture and sequencing technology identified 70 age-related differentially regulated methylation regions in 738 individuals [99].

The DNMT and TET protein families are crucial regulators of DNA methylation and demethylation processes, and their expression differs between young and old HSCs [100, 101]. Initial studies revealed that HSCs with reduced DNMT1 activity in mice could differentiate into myeloid erythrocytes but not lymphocytes, emphasizing the critical role of DNA methylation in HSC self-renewal activity [102]. Specific deletion of DNMT1 directly links DNA methylation to HSC senescence, resulting in a lineage bias toward myelopoiesis and self-renewal defects, characteristic of natural HSC senescence [103]. Wall found that as HSCs undergo differentiation into a lineage-committed state characterized by enhanced methylation of stemness genes and diminished methylation of lineage-specific genes. This suggests that aberrant activation or inactivation of demethylases or methylases may disrupt this balance, underscoring the crucial role of DNA methylases in HSC homeostasis [36, 104]. The TET demethylase is also implicated in aging. Quantitative mass spectrometry analysis revealed that 5hmC levels decrease during HSC aging in both mice and humans, indicating reduced 5hmC levels as an epigenetic mark of HSC aging [105, 106].

Histone modifications

Histone modifications play a crucial role in influencing the gene expression patterns of senescent HSCs [100]. Comprehensive genomic investigations have revealed numerous age-associated genes subject to regulation during the aging process. This includes lysine-specific demethylases, which exhibit a decline in expression levels with advancing age [107–110]. Comparative analyses between young and senescent HSCs reveal broader H3K4me3 peaks in senescent HSCs associated with HSC identity and self-renewal genes. Additionally, DNA methylation of transcription factor binding sites associated with differentiation is increased. The distribution of H3K27me3 undergoes significant changes, with numerous promoter regions displaying an increase in H3K27me3 marks [101]. Histone acetylation has also been implicated in HSC senescence. Under stress conditions, the deletion of the deacetylase SIRT1 in HSCs leads to an increase in HSC numbers, ultimately resulting in DNA damage and HSC depletion in mice [111]. Furthermore, young HSCs exhibit high levels of H4K16 acetylation, while a subset of senescent HSCs shows a significant decrease in H4K16Ac [112].

It is evident that alterations in histone modifications impact the state of HSCs, and researchers have utilized single-cell technology to delve into this relationship. In a recent study, Hui observed that BRD4 impairs HSC self-renewal and differentiation in the hematopoietic system. Transposase accessible chromatin sequencing (ATAC-seq) analysis reveals increased chromatin accessibility in Brd4Δ/Δ HSC/HPC [113]. Cheung introduced an epigenetic landscape analysis using Time-Of-Flight cytometry, a single-cell method based on mass spectrometry cytometry to analyze various histones in single cells. This approach revealed a persistent increase in cell-to-cell variability in chromatin markers in human immune cells, a hallmark of immune cell senescence. The single-cell methodology offers novel insights into the role of epigenetic regulation in hematopoiesis, immune cell function and the aging of the immune system. Additionally, it reveals aberrant epigenetic patterns linked to immune-mediated diseases [114].

Chromatin remodeling

Single-cell assay for transposase accessible chromatin sequencing (scATAC-seq) is used to analyze chromatin accessibility and detect dynamic changes in chromatin structure at a single-cell level. Using scATAC-seq, Florian et al. revealed that chromatin structure is involved in the execution of different division modes of HSCs [37]. In details, they found that young HSCs mainly undergo asymmetric division, while old HSCs mainly undergo symmetric division. Furthermore, daughter cell potential correlates with the levels of the epigenetic mark H4K16ac and the amount of open chromatin allocated to daughter cells, suggesting that epigenetic mechanisms determine the functional outcome of HSCs division.

Single-cell high-throughput chromosome conformation capture (scHi-C) is the single-cell technology currently used to explore the three-dimensional (3D) structure of chromatin at a single-cell level. There has been a lack of complete studies on the 3D chromatin conformation of cell types at various stages of the hematopoietic lineage. The main limiting factor is that existing chromatin conformation capture techniques require large numbers of cells, and there are few HSCs in the body. Chen et al. introduced a low-input tag-based Hi-C method. They used this technique to generate 3D genomic data maps of 10 cell types during the differentiation of the mouse hematopoietic lineage. The development of scHi-C will help researchers understand various biological processes in the hematopoietic system [115].

PROTEOMIC ALTERATIONS CORRELATED WITH HEMATOPOIETIC AGING

Single-cell transcriptomics have transformed our understanding of hematopoietic system by facilitating gene expression measurements at high resolution, but protein-level expression measurements in single cells provide added value over measuring RNA-level expression alone, as the relationship between them is not a straightforward correlation for most genes. A few studies applied single-cell proteomics to the research in hematopoietic system. Palii et al. used single-cell proteomic with temporal barcoding to capture the temporal dynamics of protein expression of lineage-specific transcription factors (LS-TFs) in individual cells during human erythropoiesis using, and found that LS-TFs from alternate lineages are co-expressed in individual early progenitor cells and expression of them gradually changes to direct cell-fate decisions [116]. In a recent study, Zhang identified more than 80 different subpopulations of HSPCs by performing cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) on human bone marrow cells and constructed an immunophenotype-coupled transcriptomic atlas of human hematopoietic progenitors [40]. Zhao et al. adapted a high-throughput microfluidic-based technology to quantify 15 secreted proteins in HSPCs at single-cell level, and revealed that, upon Toll-like receptor stimulation, short-term HSCs and multipotent progenitor cells can produce a large number of diverse cytokines through NF-κB signaling [117]. Single-cell proteomics by mass cytometry time of flight (CyTOF) revealed impact of genetic alterations on frequency and lineage potential of HSC, providing mechanistic insights into altered hematopoiesis at single cell resolution [118]. However, few studies applied single-cell proteomics to the research in the aging dynamics of hematopoietic system, except that one group using single-cell proteomics by CyTOF measuring 20 surface proteins identified G6B as an indicator of mutant clone-derived HSPCs in myelofibrosis, which is often associated with aging [119].

METABOLOMICS ALTERATIONS CORRELATED WITH HEMATOPOIETIC AGING

The phenotypic manifestations in aging organisms are influenced by the repercussions of aging on physiological processes. A researcher performed proteomic analysis of human HSCs at different ages and other cells within the bone marrow niche. Their results show that the metabolic and anabolic activities of senescent hematopoietic progenitor cells (HPCs) are enhanced, and the lineage bias of HSPCs toward myeloid differentiation is related to glycolysis [35]. Studies have shown that aging phenotypes can be alleviated through caloric restriction [120]. Employing single-cell transcriptomic analysis on various tissues from calorically restricted aging rats uncovered the suppressive impact of caloric restriction on age-related inflammation [121].

While early studies tentatively proposed an association between dietary restriction (DR) and amelioration of HSC aging, these were primarily confined to phenotypic observations and lacked comprehensive exploration. Tao elucidated that intermediate-onset DR rectified the perturbed equilibrium in the HSC pool in aged mice, suppressing B lymphopoiesis under unperturbed conditions. Both short- and medium-term DR exhibited improvements in hematopoietic regeneration within aging HSCs, with long-term DR showcasing enhanced repopulation abilities of total bone marrow cells. Mechanistically, DR rejuvenated dysregulated mitochondrial pathways, increased quiescence and diminished DNA damage signaling [122]. Ma conducted extensive single-cell and single-nucleus transcriptome analyses across various tissues to investigate the impact of aging and caloric restriction in rats. The findings indicate that caloric restriction mitigates age-related alterations in cell type composition, gene expression and core transcriptional regulatory networks. This research offers a comprehensive single-cell transcriptional landscape across multiple tissues associated with mammalian aging and caloric restriction. It enhances our comprehension of caloric restriction’s resilience as an age-protective intervention and elucidates the mechanisms through which metabolic interventions reshape the aging process of the immune system. [121].

The gut microbiota stands as a pivotal regulator of host health and immunity. A recent study employing fecal microbiota transplantation (FMT) from young to aged mice unveiled noteworthy lymphoid differentiation in aged recipient mice, featuring increased and reduced myeloid differentiation. FMT from young mice exerted a rejuvenating effect on aged HSCs, enhancing both short- and long-term hematopoietic repopulation capacities. Metabolomic analysis disclosed the reshaping of intestinal microbiota composition and metabolites following FMT, with Treponema and tryptophan-related metabolites emerging as key contributors to hematopoiesis recovery and the rejuvenation of aged HSCs [123].

SINGLE-CELL AND SPATIAL MULTI-OMICS ANALYSIS OF HEMATOPOIETIC AGING

Single-cell techniques have advanced our understanding of hematopoietic aging, notably revealing many changes in aging HSCs (Fig. 2). Given the intricate landscape of cellular diversity within organisms, there arises a necessity for a comprehensive capture and analysis of multiple cellular processes to unravel this complexity. The advent of single-cell multi-omics technology addresses this need by facilitating the simultaneous integration of various single-omics methods, encompassing transcriptomics, genomics, epigenomics, epitranscriptomics, proteomics, metabolomics and other omics. This integrated approach elevates our comprehension of biological mechanisms and genotype–phenotype relationships, thereby instigating a transformative impact on molecular cell biology research [13, 124]. The evolution of single-cell multi-omics technologies empowers the direct characterization of phenotypes that had to be inferred through single-omics approaches. These advanced methodologies unravel the intricacies of gene regulation mechanisms, dynamics in protein expression, variations in epigenetic landscapes and models of cell perturbation. Consequently, they offer novel insights into comprehending cellular heterogeneity in both health and disease [15, 124–127], with a particular emphasis on studies within the hematopoietic system. At present, many achievements have been made in revealing the physiological or pathological processes of the hematopoietic system through Single-cell multi-omics methods. For example, deciphering cell status and human hematopoietic lineage [128, 129], explaining the occurrence and development of various blood system diseases [130–132]. CITE-seq addresses certain challenges inherent in single-cell analysis by integrating technologies for both transcriptomic and surface proteomic profiling. This innovative approach enables the concurrent detection of cell surface protein information and intracellular transcriptome information at the single-cell level. CITE-seq enhances the depth of understanding of individual cells, providing a more nuanced grasp of cellular heterogeneity and a more precise delineation of specific cell types, particularly within the intricate hematopoietic system [133–135]. However, it is essential to note that CITE-seq is not without limitations. It necessitates simultaneous antigen detection and transcriptome sequencing, imposing stringent requirements on cell viability for the latter process. Presently, CITE-seq predominantly targets membrane surface antigens to avoid potential impacts on the transcriptome when antibodies permeate the membrane. The extension of proteomic analysis tools using antibodies beyond cell surface proteins remains an area warranting further exploration and development.

Overview of the changes in aging HSCs revealed by single-cell technology. Compared with young HSCs, old HSCs exhibit asymmetric division, reduced G1 phase cell number, increased epigenetic reprogramming and up-regulated expression of Nupr1, Vwf, Clu and other proteins. Treponema and tryptophan-related metabolites emerge as key contributors to hematopoiesis recovery and the rejuvenation of aged HSCs
Figure 2

Overview of the changes in aging HSCs revealed by single-cell technology. Compared with young HSCs, old HSCs exhibit asymmetric division, reduced G1 phase cell number, increased epigenetic reprogramming and up-regulated expression of Nupr1, Vwf, Clu and other proteins. Treponema and tryptophan-related metabolites emerge as key contributors to hematopoiesis recovery and the rejuvenation of aged HSCs

The principal constraint of single-cell methodologies is rooted in their reliance on cell suspensions or isolated nuclei, necessitating subsequent disaggregation before analysis and consequently resulting in the forfeiture of crucial spatial information. The escalating demand for location-specific insights has spurred the development of spatiomics. As single-cell technology progresses, the capacity to investigate diverse molecular analytes within their native tissue context at subcellular resolution has given rise to the concept of ‘spatial multi-omics,’ acknowledged as a noteworthy technology by Nature magazine in 2022 [136]. Researches indicate that, beyond intrinsic factors, certain extrinsic elements can influence HSC aging, with the bone marrow microenvironment being a critical determinant of HSC function [137–145]. Moreover, non-hematopoietic cells play a crucial role in the HSC niche [146–148]. Spatiomics has proven valuable in diverse fields, yielding insights into development and brain structure [149–152], tumors [153–156] and neurodegenerative diseases [157, 158]. Hence, spatial multi-omics holds promise for elucidating the interplay between the aging and functional alterations of HSCs and the bone marrow milieu. Despite being in its nascent stages, spatial omics is advancing swiftly, and these nascent methodologies are poised to enhance our profound comprehension of biology [159].

FUTURE

Since Tang [11] pioneered the development of the world’s first single-cell sequencing technology in 2009, the field of single-cell technology has experienced rapid growth and widespread application over the past decade. Evolving from its initial capacity to analyze only a limited number of cells, the technology has progressed to simultaneously assess millions of cells. However, persistent challenges arise in ascertaining cellular origin and subsequent differentiation due to inherent technical constraints. To surmount these challenges, lineage tracing, at the forefront of technology, intervenes by imprinting cells with stable genetic markers, facilitating the tracking of cellular processes such as division, differentiation, migration and more. Wang have introduced a pioneering lineage tracing mouse model, DARLIN, consequently spearheading the development of Camallia-seq, the world’s premier single-cell multi-omics lineage tracing technology. This groundbreaking methodology allows for the concurrent detection of lineage markers, transcriptomic profiles, global DNA methylation and chromatin accessibility at the single-cell level. These innovations have proven pivotal in scrutinizing the fate selection of HSCs, their migration across diverse bone marrow microenvironments and the perpetuation of cellular molecular imprints. The significance of these advancements transcends individual studies, furnishing life sciences researchers with a potent tool to surmount challenges within their respective domains [160].

The landscape of technology is in constant flux to meet evolving demands, and the realm of single-cell technology is no exception, experiencing continuous innovation. Specifically, second-generation sequencing-based scWGS has the capability to interrogate genetic variations at the whole-genome level at the single-cell resolution. However, the relatively short sequencing read length associated with scWGS introduces certain limitations. In contrast, third-generation sequencing (TGS) technology presents notable advantages in examining the transcriptome of individual cells. The strengths of TGS technology lie in its long-read lengths, elimination of the need for amplification, facilitation of direct sequencing, and minimal input requirements. TGS technology has emerged as an indispensable tool for single-cell analysis, providing more exhaustive, accurate, and profound single-cell transcriptome data.

Despite the widespread adoption of single-cell omics in both basic research and clinical trials, several notable challenges persist. The preparation of single-cell samples typically entails compromises in terms of cell integrity and activity. The enhancement of preparation methods holds the potential to improve both cell throughput and accuracy. Determining the optimal sequencing depth is another intricate challenge, as heightened depth escalates sequencing costs. Achieving a judicious equilibrium between throughput, cost considerations, and the intricacies of data processing is imperative. This entails a comprehensive evaluation of research objectives, available resources and pertinent technical attributes.

To comprehensively elucidate the molecular hierarchy from the genome to phenotype within a single cell, the integration of multi-omics approaches with both single-cell and spatial resolution becomes imperative. These techniques enable a clear exploration of the intermolecular dynamics between gene regulation and gene expression within the same cell type, particularly in investigations pertaining to development, aging and disease. Furthermore, these advanced technologies empower the investigation of how acquired genetic variations in single-cell genomes influence their functional and phenotypic characteristics, as well as the functions of surrounding tissues. This not only provides avenues for a broad range of applications in cell biology but also offers unprecedented insights into the intricacies of biological processes. The ongoing progress in single-cell technology and the convergence of multi-omics with spatial resolution are poised to drive further innovation, promising even more comprehensive insights into our understanding of biology in the future.

Key Points
  • Single-Cell Revolution: Explores how high-resolution single-cell sequencing has revolutionized our understanding of hematopoietic aging by eliminating heterogeneity to a significant extent.

  • Molecular Dynamics: Investigates the intermolecular dynamics between gene regulation and expression within the same cell type, providing a nuanced view of the aging process at the molecular level.

  • Temporal Dynamics: Highlights the urgency and necessity of high-resolution analyses to decipher the real-time, quantitative dynamics of HSCs during aging.

ACKNOWLEDGEMENTS

This work was supported by the National Key R&D Program of China (2021YFA1102800) and by National Natural Science Foundation of China (92249304, 82301746). This work was supported by the Science Foundation for Distinguished Young Scholars of Guangdong Province (2019B151502008) to Hu Wang. This work was also supported by the Hangzhou Youth Innovation Team Project (TD2023020).

AUTHOR CONTRIBUTIONS

Xinrong Jin (Investigation [equal], Software [equal], Visualization [equal], Writing— original draft [lead), Ruohan Zhang (Investigation [equal], Writing—original draft [supporting]), Yunqi Fu (Investigation [equal], Writing—original draft [supporting]), Qiunan Zhu (Investigation [equal], Writing—original draft-Supporting), Liquan Hong (Conceptualization [equal], Methodology [equal], Project administration [equal], Resources [equal], Supervision [equal], Writing—review & editing [equal]), Aiwei Wu (Software [equal], Supervision [equal], Visualization [equal], Writing—review & editing [equal]), Hu Wang (Conceptualization [equal], Funding acquisition [equal], Project administration [equal], Resources [equal], Supervision [equal], Writing—review & editing [equal]).

Author Biographies

Xinrong Jin is currently pursuing his Ph.D. in Biology from Hangzhou Normal University under the supervision of Hu Wang. His research interests focus on the mechanism of stem cell aging.

Ruohan Zhang is an undergraduate student at the Basic Medical Sciences at Hangzhou Normal University. Her research interests focus on mechanism of stem cell aging.

Yunqi Fu is an undergraduate student at the Basic Medical Sciences at Hangzhou Normal University. His research interests focus on machine learning for data processing.

Qiunan Zhu is an undergraduate student at the Basic Medical Sciences at Hangzhou Normal University. His research interests focus on the mechanism of mouse adipocytes aging.

Liquan Hong, is the director of the Third People’s Hospital of Deqing, China. His research interests focus on the mechanism of stem cell aging.

Aiwei Wu earned her Ph.D. from Shanghai Jiao Tong University. Her research interests lie in understanding the mechanisms of hematopoietic aging utilizing cutting-edge single-cell multiomics and bioinformatics approaches.

Hu Wang earned his Ph.D. from Zhejiang University. He is currently dedicated to investigating the molecular mechanisms of adult stem cell aging, as well as advancing the development and application of anti-aging drugs.

References

1.

Liggett
LA
,
Sankaran
VG
.
Unraveling Hematopoiesis through the lens of genomics
.
Cell
 
2020
;
182
:
1384
400
.

2.

Laurenti
E
,
Göttgens
B
.
From haematopoietic stem cells to complex differentiation landscapes
.
Nature
 
2018
;
553
:
418
26
.

3.

Vaziri
H
,
Dragowska
W
,
Allsopp
RC
, et al.  
Evidence for a mitotic clock in human hematopoietic stem cells: loss of telomeric DNA with age
.
Proc Natl Acad Sci U S A
 
1994
;
91
:
9857
60
.

4.

Pang
WW
,
Price
EA
,
Sahoo
D
, et al.  
Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age
.
Proc Natl Acad Sci U S A
 
2011
;
108
:
20012
7
.

5.

Nilsson
AR
,
Soneji
S
,
Adolfsson
S
, et al.  
Human and murine hematopoietic stem cell aging is associated with functional impairments and intrinsic megakaryocytic/erythroid bias
.
PloS One
 
2016
;
11
:
11
.

6.

Morrison
SJ
,
Wandycz
AM
,
Akashi
K
, et al.  
The aging of hematopoietic stem cells
.
Nat Med
 
1996
;
2
:
1011
6
.

7.

Yamashita
M
,
Dellorusso
PV
,
Olson
OC
,
Passegué
E
.
Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis
.
Nat Rev Cancer
 
2020
;
20
:
365
82
.

8.

Díaz
PAC
,
Mistry
JJ
,
Trowbridge
JJ
.
Hematopoietic stem cell aging and leukemia transformation
.
Blood
 
2023
;
142
:
533
42
.

9.

Denkinger
MD
,
Leins
H
,
Schirmbeck
R
, et al.  
HSC aging and senescent immune Remodeling
.
Trends Immunol
 
2015
;
36
:
815
24
.

10.

Eberwine
J
,
Yeh
H
,
Miyashiro
K
, et al.  
Analysis of gene expression in single live neurons
.
Proc Natl Acad Sci U S A
 
1992
;
89
:
3010
4
.

11.

Tang
FC
,
Barbacioru
C
,
Wang
YZ
, et al.  
mRNA-Seq whole-transcriptome analysis of a single cell
.
Nat Methods
 
2009
;
6
:
377
82
.

12.

Jovic
D
,
Liang
X
,
Zeng
H
, et al.  
Single-cell RNA sequencing technologies and applications: a brief overview
.
Clin Transl Med
 
2022
;
12
:e694.

13.

Lee
J
,
Hyeon
DY
,
Hwang
D
.
Single-cell multiomics: technologies and data analysis methods
.
Exp Mol Med
 
2020
;
52
:
1428
42
.

14.

Lei
Y
,
Tang
R
,
Xu
J
, et al.  
Applications of single-cell sequencing in cancer research: progress and perspectives
.
J Hematol Oncol
 
2021
;
14
:
91
.

15.

He
X
,
Memczak
S
,
Qu
J
, et al.  
Single-cell omics in ageing: a young and growing field
.
Nat Metab
 
2020
;
2
:
293
302
.

16.

Xu
XJ
,
Hua
XM
,
Mo
H
, et al.  
Single-cell RNA sequencing to identify cellular heterogeneity and targets in cardiovascular diseases: from bench to bedside
.
Basic Res Cardiol
 
2023
;
118
:
7
.

17.

Allen
WE
,
Blosser
TR
,
Sullivan
ZA
, et al.  
Molecular and spatial signatures of mouse brain aging at single-cell resolution
.
Cell
 
2023
;
186
:
194
,
194
208 e118
,
208.e18
.

18.

Rahimzadeh
N
,
Srinivasan
SS
,
Zhang
J
,
Swarup
V
.
Gene networks and systems biology in Alzheimer's disease: insights from multi-omics approaches
.
Alzheimers Dement
 
2024
.

19.

Yang
JW
,
Zhou
X
,
Dong
J
, et al.  
Single-cell profiling reveals molecular basis of malignant phenotypes and tumor microenvironments in small bowel adenocarcinomas
.
Cell Discovery
 
2022
;
8
:
8
.

20.

Guo
LW
,
Wang
YJ
,
Yang
WX
, et al.  
Molecular profiling provides clinical insights into targeted and immunotherapies as well as colorectal cancer prognosis
.
Gastroenterology
 
2023
;
165
.

21.

Alonso-Curbelo
D
,
Ho
YJ
,
Burdziak
C
, et al.  
A gene-environment-induced epigenetic program initiates tumorigenesis
.
Nature
 
2021
;
590
:
642
8
.

22.

Sun
B
,
Xun
Z
,
Zhang
N
, et al.  
Single-cell RNA sequencing in cancer research: discovering novel biomarkers and therapeutic targets for immune checkpoint blockade
.
Cancer Cell Int
 
2023
;
23
:
313
.

23.

Rodriguez-Meira
A
,
Buck
G
,
Clark
SA
, et al.  
Unravelling Intratumoral heterogeneity through high-sensitivity single-cell mutational analysis and parallel RNA sequencing
.
Mol Cell
 
2019
;
73
:
1292
1305.e8
.

24.

Nam
AS
,
Kim
KT
,
Chaligne
R
, et al.  
Somatic mutations and cell identity linked by genotyping of transcriptomes
.
Nature
 
2019
;
571
:
355
60
.

25.

Acha
P
,
Palomo
L
,
Fuster-Tormo
F
, et al.  
Analysis of Intratumoral heterogeneity in myelodysplastic syndromes with isolated del(5q) using a single cell approach
.
Cancer
 
2021
;
13
.

26.

Fan
J
,
Wang
LL
,
Francis
JM
, et al.  
Integrated analysis of targeted single-cell genetic and transcriptional heterogeneity suggests novel drivers of chronic lymphocytic leukemia
.
Clin Cancer Res
 
2017
;
23
:
27
51
.

27.

Guo
XX
,
Xu
WL
,
Zhang
WP
, et al.  
High-frequency and functional mitochondrial DNA mutations at the single-cell level
.
Proc Natl Acad Sci U S A
 
2022
;
120
.

28.

Kowalczyk
MS
,
Tirosh
I
,
Heckl
D
, et al.  
Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells
.
Genome Res
 
2015
;
25
:
1860
72
.

29.

Janzen
V
,
Forkert
R
,
Fleming
HE
, et al.  
Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a
.
Nature
 
2006
;
443
:
421
6
.

30.

Grover
A
,
Sanjuan-Pla
A
,
Thongjuea
S
, et al.  
Single-cell RNA sequencing reveals molecular and functional platelet bias of aged haematopoietic stem cells
.
Nat Commun
 
2016
;
7
:
11075
.

31.

Herault
L
,
Poplineau
M
,
Mazuel
A
, et al.  
Single-cell RNA-seq reveals a concomitant delay in differentiation and cell cycle of aged hematopoietic stem cells
.
BMC Biol
 
2021
;
19
:
19
.

32.

Mincarelli
L
,
Uzun
V
,
Wright
D
, et al.  
Single-cell gene and isoform expression analysis reveals signatures of ageing in haematopoietic stem and progenitor cells
.
Commun Biol
 
2023
;
6
:
558
.

33.

Adelman
ER
,
Huang
HT
,
Roisman
A
, et al.  
Aging human hematopoietic stem cells manifest profound epigenetic reprogramming of enhancers that may predispose to Leukemia
.
Cancer Discov
 
2019
;
9
:
1080
101
.

34.

Ma
JL
,
Pettit
N
,
Talburt
J
, et al.  
Integrating single-cell transcriptome and network analysis to characterize the therapeutic response of chronic myeloid Leukemia
.
Int J Mol Sci
 
2022
;
23
:
23
.

35.

Hennrich
ML
,
Romanov
N
,
Horn
P
, et al.  
Cell-specific proteome analyses of human bone marrow reveal molecular features of age-dependent functional decline
.
Nat Commun
 
2018
;
9
:
4004
.

36.

Jianga
PL
,
Wanga
H
,
Zhenga
JC
, et al.  
Epigenetic regulation of hematopoietic stem cell homeostasis
.
Blood Science
 
2019
;
1
:
19
28
.

37.

Florian
MC
,
Klose
M
,
Sacma
M
, et al.  
Aging alters the epigenetic asymmetry of HSC division
.
PLoS Biol
 
2018
;
16
:
e2003389
.

38.

Hui
T
,
Cao
Q
,
Wegrzyn-Woltosz
J
, et al.  
High-resolution single-cell DNA methylation measurements reveal epigenetically distinct hematopoietic stem cell subpopulations
.
Stem Cell Reports
 
2018
;
11
:
578
92
.

39.

Rodriguez-Meira
A
,
O'Sullivan
J
,
Rahman
H
, et al.  
TARGET-Seq: a protocol for high-sensitivity single-cell mutational analysis and parallel RNA sequencing
.
STAR Protoc
 
2020
;
1
:
100125
.

40.

Zhang
X
,
Song
B
,
Carlino
MJ
, et al.  
An immunophenotype-coupled transcriptomic atlas of human hematopoietic progenitors
.
Nat Immunol
 
2024
;
25
:
703
15
.

41.

Zhao
Y
,
Simon
M
,
Seluanov
A
,
Gorbunova
V
.
DNA damage and repair in age-related inflammation
.
Nat Rev Immunol
 
2023
;
23
:
75
89
.

42.

Hoang
ML
,
Kinde
I
,
Tomasetti
C
, et al.  
Genome-wide quantification of rare somatic mutations in normal human tissues using massively parallel sequencing
.
Proc Natl Acad Sci U S A
 
2016
;
113
:
9846
51
.

43.

Welch
JS
,
Ley
TJ
,
Link
DC
, et al.  
The origin and evolution of mutations in acute myeloid leukemia
.
Cell
 
2012
;
150
:
264
78
.

44.

Jaiswal
S
,
Fontanillas
P
,
Flannick
J
, et al.  
Age-related clonal hematopoiesis associated with adverse outcomes
.
N Engl J Med
 
2014
;
371
:
2488
98
.

45.

Genovese
G
,
Kahler
AK
,
Handsaker
RE
, et al.  
Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence
.
N Engl J Med
 
2014
;
371
:
2477
87
.

46.

Steensma
DP
.
Clinical consequences of clonal hematopoiesis of indeterminate potential
.
Blood Adv
 
2018
;
2
:
3404
10
.

47.

Lim
J
,
Chin
V
,
Fairfax
K
, et al.  
Transitioning single-cell genomics into the clinic
.
Nat Rev Genet
 
2023
;
24
:
573
84
.

48.

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
:
1622
6
.

49.

Dey
SS
,
Kester
L
,
Spanjaard
B
, et al.  
Integrated genome and transcriptome sequencing of the same cell
.
Nat Biotechnol
 
2015
;
33
:
285
9
.

50.

Macaulay
IC
,
Haerty
W
,
Kumar
P
, et al.  
G&T-seq: parallel sequencing of single-cell genomes and transcriptomes
.
Nat Methods
 
2015
;
12
:
519
22
.

51.

Zachariadis
V
,
Cheng
H
,
Andrews
N
, et al.  
A highly scalable method for joint whole-genome sequencing and gene-expression profiling of single cells
.
Mol Cell
 
2020
;
80
:
541
553 e545
.

52.

Strzelecka
PM
,
Damm
F
.
Haematopoietic ageing through the lens of single-cell technologies
.
Dis Model Mech
 
2021
;
14
.

53.

De Bie
J
,
Demeyer
S
,
Alberti-Servera
L
, et al.  
Single-cell sequencing reveals the origin and the order of mutation acquisition in T-cell acute lymphoblastic leukemia
.
Leukemia
 
2018
;
32
:
1358
69
.

54.

Alberti-Servera
L
,
Demeyer
S
,
Govaerts
I
, et al.  
Single-cell DNA amplicon sequencing reveals clonal heterogeneity and evolution in T-cell acute lymphoblastic leukemia
.
Blood
 
2021
;
137
:
801
11
.

55.

Gawad
C
,
Koh
W
,
Quake
SR
.
Dissecting the clonal origins of childhood acute lymphoblastic leukemia by single-cell genomics
.
Proc Natl Acad Sci U S A
 
2014
;
111
:
17947
52
.

56.

Popov
LD
.
Mitochondria as intracellular signalling organelles. An update
.
Cell Signal
 
2023
;
109
:
110794
.

57.

Kazak
L
,
Reyes
A
,
Holt
IJ
.
Minimizing the damage: repair pathways keep mitochondrial DNA intact
.
Nat Rev Mol Cell Biol
 
2012
;
13
:
659
71
.

58.

Kauppila
TES
,
Kauppila
JHK
,
Larsson
NG
.
Mammalian mitochondria and aging: An update
.
Cell Metab
 
2017
;
25
:
57
71
.

59.

Zhang
HB
,
Menzies
KJ
,
Auwerx
J
.
The role of mitochondria in stem cell fate and aging
.
Development
 
2018
;
145
:
145
.

60.

Sevini
F
,
Giuliani
C
,
Vianello
D
, et al.  
mtDNA mutations in human aging and longevity: controversies and new perspectives opened by high-throughput technologies
.
Exp Gerontol
 
2014
;
56
:
234
44
.

61.

Kopinski
PK
,
Singh
LN
,
Zhang
SP
, et al.  
Mitochondrial DNA variation and cancer
.
Nat Rev Cancer
 
2021
;
21
:
431
45
.

62.

Otten
ABC
,
Sallevelt
SCEH
,
Carling
PJ
, et al.  
Mutation-specific effects in germline transmission of pathogenic mtDNA variants
.
Hum Reprod
 
2018
;
33
:
1331
41
.

63.

Ye
KX
,
Lu
J
,
Ma
F
, et al.  
Extensive pathogenicity of mitochondrial heteroplasmy in healthy human individuals
.
Proc Natl Acad Sci U S A
 
2014
;
111
:
10654
9
.

64.

Sanchez-Contreras
M
,
Kennedy
SR
.
The complicated nature of somatic mtDNA mutations in aging, front
.
Aging
 
2022
;
2
.

65.

Morris
J
,
Na
YJ
,
Zhu
H
, et al.  
Pervasive within-mitochondrion single-nucleotide variant Heteroplasmy as revealed by single-mitochondrion sequencing
.
Cell Rep
 
2017
;
21
:
2706
13
.

66.

Ludwig
LS
,
Lareau
CA
,
Ulirsch
JC
, et al.  
Lineage tracing in humans enabled by mitochondrial mutations and single-cell genomics
.
Cell
 
2019
;
176
:
1325
1339.e22
.

67.

Xu
J
,
Nuno
K
,
Litzenburger
UM
, et al.  
Single-cell lineage tracing by endogenous mutations enriched in transposase accessible mitochondrial DNA
.
Elife
 
2019
;
8
:
8
.

68.

Lareau
CA
,
Ludwig
LS
,
Muus
C
, et al.  
Massively parallel single-cell mitochondrial DNA genotyping and chromatin profiling (vol 39, pg 451, 2021)
.
Nat Biotechnol
 
2023
;
41
:
1345
.

69.

Lareau
CA
,
Liu
VC
,
Muus
C
, et al.  
Mitochondrial single-cell ATAC-seq for high-throughput multi-omic detection of mitochondrial genotypes and chromatin accessibility
.
Nat Protoc
 
2023
;
18
:
1416
40
.

70.

Liyanage
SU
,
Hurren
R
,
Voisin
V
, et al.  
Leveraging increased cytoplasmic nucleoside kinase activity to target mtDNA and oxidative phosphorylation in AML
.
Blood
 
2017
;
129
:
2657
66
.

71.

Blasiak
J
,
Hoser
G
,
Bialkowska-Warzecha
J
, et al.  
Reactive oxygen species and mitochondrial DNA damage and repair in BCR-ABL1 cells resistant to Imatinib
.
Biores Open Access
 
2015
;
4
:
334
42
.

72.

Flohr Svendsen
A
,
Yang
D
,
Kim
K
, et al.  
A comprehensive transcriptome signature of murine hematopoietic stem cell aging
.
Blood
 
2021
;
138
:
439
51
.

73.

Kirschner
K
,
Chandra
T
,
Kiselev
V
, et al.  
Proliferation drives aging-related functional decline in a subpopulation of the hematopoietic stem cell compartment
.
Cell Rep
 
2017
;
19
:
1503
11
.

74.

Oetjen
KA
,
Lindblad
KE
,
Goswami
M
, et al.  
Human bone marrow assessment by single-cell RNA sequencing, mass cytometry, and flow cytometry, JCI
.
Insight
 
2018
;
3
:
3
.

75.

Kucinski
I
,
Campos
J
,
Barile
M
, et al.  
A time- and single-cell-resolved model of murine bone marrow hematopoiesis
.
Cell Stem Cell
 
2023
.

76.

Tabula
MC
.
A single-cell transcriptomic atlas characterizes ageing tissues in the mouse
.
Nature
 
2020
;
583
:
590
5
.

77.

Zheng
YF
,
Liu
XX
,
Le
WQ
, et al.  
A human circulating immune cell landscape in aging and COVID-19
.
Protein Cell
 
2020
;
11
:
740
70
.

78.

Arthur
L
,
Esaulova
E
,
Mogilenko
DA
, et al.  
Cellular and plasma proteomic determinants of COVID-19 and non-COVID-19 pulmonary diseases relative to healthy aging
.
Nature Aging
 
2021
;
1
:
535
49
.

79.

Hashimoto
K
,
Kouno
T
,
Ikawa
T
, et al.  
Single-cell transcriptomics reveals expansion of cytotoxic CD4 T cells in supercentenarians
.
Proc Natl Acad Sci U S A
 
2019
;
116
:
24242
51
.

80.

Schaum
N
,
Lehallier
B
,
Hahn
O
, et al.  
Ageing hallmarks exhibit organ-specific temporal signatures
.
Nature
 
2020
;
583
:
596
602
.

81.

Jaiswal
S
,
Ebert
BL
.
Clonal hematopoiesis in human aging and disease
.
Science
 
2019
;
366
:
366
.

82.

Giustacchini
A
,
Thongjuea
S
,
Barkas
N
, et al.  
Single-cell transcriptomics uncovers distinct molecular signatures of stem cells in chronic myeloid leukemia
.
Nat Med
 
2017
;
23
:
692
702
.

83.

Zhang
WL
,
Yang
BB
,
Weng
LQ
, et al.  
Single cell sequencing reveals cell populations that predict primary resistance to imatinib in chronic myeloid leukemia
.
Aging-Us
 
2020
;
12
:
25337
55
.

84.

Krishnan
V
,
Schmidt
F
,
Nawaz
Z
, et al.  
A single-cell atlas identifies pretreatment features of primary imatinib resistance in chronic myeloid leukemia
.
Blood
 
2023
;
141
:
2738
55
.

85.

Wang
LL
,
Fan
J
,
Francis
JM
, et al.  
Integrated single-cell genetic and transcriptional analysis suggests novel drivers of chronic lymphocytic leukemia
.
Genome Res
 
2017
;
27
:
1300
11
.

86.

van
 
Galen
P
,
Hovestadt
V
,
Wadsworth
MH
, et al.  
Single-cell RNA-Seq reveals AML hierarchies relevant to disease progression and immunity
.
Cell
 
2019
;
176
:
1265
1281.e24
.

87.

Lohr
JG
,
Kim
S
,
Gould
J
, et al.  
Genetic interrogation of circulating multiple myeloma cells at single-cell resolution
.
Sci Transl Med
 
2016
;
8
:
8
.

88.

Tong
JY
,
Sun
T
,
Ma
SH
, et al.  
Hematopoietic stem cell heterogeneity is linked to the initiation and therapeutic response of myeloproliferative neoplasms
.
Cell Stem Cell
 
2021
;
28
:
502
513.e6
.

89.

Saul
D
,
Kosinsky
RL
.
Epigenetics of aging and aging-associated diseases
.
Int J Mol Sci
 
2021
;
22
:
22
.

90.

Raghuwanshi
S
,
Dahariya
S
,
Kandi
R
, et al.  
Epigenetic mechanisms: role in hematopoietic stem cell lineage commitment and differentiation
.
Curr Drug Targets
 
2018
;
19
:
1683
95
.

91.

López-Otin
C
,
Blasco
MA
,
Partridge
L
, et al.  
Hallmarks of aging: An expanding universe
.
Cell
 
2023
;
186
:
243
78
.

92.

Mejia-Ramirez
E
,
Florian
MC
.
Understanding intrinsic hematopoietic stem cell aging
.
Haematologica
 
2020
;
105
:
22
37
.

93.

Wang
K
,
Liu
HC
,
Hu
QC
, et al.  
Epigenetic regulation of aging: implications for interventions of aging and diseases, signal transduction and targeted
.
Therapy
 
2022
;
7
:
7
.

94.

Luo
CY
,
Hajkova
P
,
Ecker
JR
.
Dynamic DNA methylation: In the right place at the right time
.
Science
 
2018
;
361
:
1336
40
.

95.

Skvortsova
K
,
Iovino
N
,
Bogdanovic
O
.
Functions and mechanisms of epigenetic inheritance in animals
.
Nat Rev Mol Cell Biol
 
2018
;
19
:
774
90
.

96.

Krepelova
A
,
Neri
F
.
DNA methylation controls hematopoietic stem cell aging
.
Nature Aging
 
2023
;
3
:
1320
2
.

97.

Laird
PW
.
Principles and challenges of genomewide DNA methylation analysis
.
Nat Rev Genet
 
2010
;
11
:
191
203
.

98.

Lovkvist
C
,
Dodd
IB
,
Sneppen
K
, et al.  
DNA methylation in human epigenomes depends on local topology of CpG sites
.
Nucleic Acids Res
 
2016
;
44
:
5123
32
.

99.

McClay
JL
,
Aberg
KA
,
Clark
SL
, et al.  
A methylome-wide study of aging using massively parallel sequencing of the methyl-CpG-enriched genomic fraction from blood in over 700 subjects
.
Hum Mol Genet
 
2014
;
23
:
1175
85
.

100.

Beerman
I
,
Bock
C
,
Garrison
BS
, et al.  
Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging
.
Cell Stem Cell
 
2013
;
12
:
413
25
.

101.

Sun
DQ
,
Luo
M
,
Jeong
M
, et al.  
Epigenomic profiling of Young and aged HSCs reveals concerted changes during aging that reinforce self-renewal
.
Cell Stem Cell
 
2014
;
14
:
673
88
.

102.

Bröske
AM
,
Vockentanz
L
,
Kharazi
S
, et al.  
DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction
.
Nat Genet
 
2009
;
41
:
1207
15
.

103.

Trowbridge
JJ
,
Snow
JW
,
Kim
J
,
Orkin
SH
.
DNA Methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells
.
Blood
 
2009
;
114
:
392
163
.

104.

Cabezas-Wallscheid
N
,
Klimmeck
D
,
Hansson
J
, et al.  
Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA Methylome analysis
.
Cell Stem Cell
 
2014
;
15
:
507
22
.

105.

Ko
M
,
Bandukwala
HS
,
An
J
, et al.  
Ten-eleven-translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice
.
Proc Natl Acad Sci U S A
 
2011
;
108
:
14566
71
.

106.

Li
Z
,
Cai
XQ
,
Cai
CL
, et al.  
Deletion of in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies
.
Blood
 
2011
;
118
:
4509
18
.

107.

Herzog
M
,
Josseaux
E
,
Dedeurwaerder
S
, et al.  
The histone demethylase Kdm3a is essential to progression through differentiation
.
Nucleic Acids Res
 
2012
;
40
:
7219
32
.

108.

Kidder
BL
,
Hu
GQ
,
Zhao
K
.
KDM5B focuses H3K4 methylation near promoters and enhancers during embryonic stem cell self-renewal and differentiation
.
Genome Biol
 
2014
;
15
:
R32
.

109.

Thieme
S
,
Gyárfás
T
,
Richter
C
, et al.  
The histone demethylase UTX regulates stem cell migration and hematopoiesis
.
Blood
 
2013
;
121
:
2462
73
.

110.

Cellot
S
,
Hope
KJ
,
Chagraoui
J
, et al.  
RNAi screen identifies Jarid1b as a major regulator of mouse HSC activity
.
Blood
 
2013
;
122
:
1545
55
.

111.

Singh
SK
,
Williams
CA
,
Klarmann
K
, et al.  
Sirt1 ablation promotes stress-induced loss of epigenetic and genomic hematopoietic stem and progenitor cell maintenance
.
Journal of Experimental Medicine
 
2013
;
210
:
987
1001
.

112.

Krishnan
V
,
Chow
MZY
,
Wang
ZM
, et al.  
Histone H4 lysine 16 hypoacetylation is associated with defective DNA repair and premature senescence in Zmpste24-deficient mice
.
Proc Natl Acad Sci U S A
 
2011
;
108
:
12325
30
.

113.

Yang
H
,
Sui
P
,
Guo
Y
, et al.  
Loss of BRD4 induces cell senescence in HSC/HPCs by deregulating histone H3 clipping
.
EMBO Rep
 
2023
;
24
:e57032.

114.

Cheung
P
,
Vallania
F
,
Dvorak
M
, et al.  
Single-cell epigenetics - chromatin modification atlas unveiled by mass cytometry
.
Clin Immunol
 
2018
;
196
:
40
8
.

115.

Zhang
C
,
Xu
Z
,
Yang
S
, et al.  
tagHi-C reveals 3D chromatin architecture dynamics during mouse Hematopoiesis
.
Cell Rep
 
2020
;
32
:108206.

116.

Palii
CG
,
Cheng
Q
,
Gillespie
MA
, et al.  
Single-cell proteomics reveal that quantitative changes in Co-expressed lineage-specific transcription factors determine cell fate
.
Cell Stem Cell
 
2019
;
24
:
812
820.e5
.

117.

Zhao
JL
,
Ma
C
,
O'Connell
R
, et al.  
Conversion of danger signals into cytokine signals by hematopoietic stem and progenitor cells for regulation of stress-induced Hematopoiesis
.
Blood
 
2014
;
124
:
2916
.

118.

Karra
L
,
Finger
AM
,
Shechtman
L
, et al.  
Single cell proteomics characterization of bone marrow hematopoiesis with distinct Ras pathway lesions
bioRxiv
.
2023
.

119.

Psaila
B
,
Wang
G
,
Rodriguez-Meira
A
, et al.  
Single-cell analyses reveal megakaryocyte-biased Hematopoiesis in Myelofibrosis and identify mutant clone-specific targets
.
Mol Cell
 
2020
;
78
:
477
,
477
492 e478
,
492.e8
.

120.

Fontana
L
,
Partridge
L
.
Promoting health and longevity through diet: from model organisms to humans
.
Cell
 
2015
;
161
:
106
18
.

121.

Ma
S
,
Sun
SH
,
Geng
LL
, et al.  
Caloric restriction reprograms the single-cell transcriptional landscape of aging
.
Cell
 
2020
;
180
:
984
1001.e22
.

122.

Tao
S
,
Wang
Y
,
Wu
J
, et al.  
Long-term mid-onset dietary restriction rejuvenates hematopoietic stem cells and improves regeneration capacity of total bone marrow from aged mice
.
Aging Cell
 
2020
;
19
:e13241.

123.

Zeng
X
,
Li
X
,
Li
X
, et al.  
Fecal microbiota transplantation from young mice rejuvenates aged hematopoietic stem cells by suppressing inflammation
.
Blood
 
2023
;
141
:
1691
707
.

124.

Baysoy
A
,
Bai
ZL
,
Satija
R
,
Fan
R
.
The technological landscape and applications of single-cell multi-omics
.
Nat Rev Mol Cell Biol
 
2023
;
24
:
695
713
.

125.

Vandereyken
K
,
Sifrim
A
,
Thienpont
B
,
Voet
T
.
Methods and applications for single-cell and spatial multi-omics
.
Nat Rev Genet
 
2023
;
24
:
494
515
.

126.

Wagner
DE
,
Klein
AM
.
Lineage tracing meets single-cell omics: opportunities and challenges
.
Nat Rev Genet
 
2020
;
21
:
410
27
.

127.

Zeng
Q
,
Mousa
M
,
Nadukkandy
AS
, et al.  
Understanding tumour endothelial cell heterogeneity and function from single-cell omics
.
Nat Rev Cancer
 
2023
;
23
:
544
64
.

128.

Weng
C
,
Yu
F
,
Yang
D
, et al.  
Deciphering cell states and genealogies of human haematopoiesis
.
Nature
 
2024
;
627
:
389
98
.

129.

Xia
J
,
Liu
M
,
Zhu
C
, et al.  
Activation of lineage competence in hemogenic endothelium precedes the formation of hematopoietic stem cell heterogeneity
.
Cell Res
 
2023
;
33
:
448
63
.

130.

Guo
R
,
Kong
J
,
Tang
P
, et al.  
Unbiased single-cell sequencing of hematopoietic and immune cells from aplastic Anemia reveals the contributors of Hematopoiesis failure and dysfunctional immune regulation
.
Adv Sci (Weinh)
 
2024
;
11
:
e2304539
.

131.

Rodriguez-Meira
A
,
Rahman
H
,
Norfo
R
, et al.  
Single-cell multi-omics reveals the genetic, cellular and molecular landscape of mutated leukemic transformation in MPN
.
Blood
 
2021
;
138
:
3
.

132.

Xu
JD
,
Wang
Y
,
Wei
Z
, et al.  
Single-cell transcriptomes combining with consecutive genomics reveal clonal evolution and gene regulatory networks in relapsed and refractory multiple myeloma
.
Frontiers in Cell and Developmental Biology
 
2022
;
9
.

133.

Jang
G
,
Contreras Castillo
S
,
Esteva
E
, et al.  
Stem cell decoupling underlies impaired lymphoid development during aging
.
Proc Natl Acad Sci U S A
 
2023
;
120
:e2302019120.

134.

Konturek-Ciesla
A
,
Dhapola
P
,
Zhang
Q
, et al.  
Temporal multimodal single-cell profiling of native hematopoiesis illuminates altered differentiation trajectories with age
.
Cell Rep
 
2023
;
42
:112304.

135.

Sommarin
MNE
,
Olofzon
R
,
Palo
S
, et al.  
Single-cell multiomics of human fetal hematopoiesis define a developmental-specific population and a fetal signature
.
Blood Adv
 
2023
;
7
:
5325
40
.

136.

Eisenstein
M
.
Seven technologies to watch in 2022
.
Nature
 
2022
;
601
:
658
61
.

137.

Ergen
AV
,
Boles
NC
,
Goodell
MA
.
Rantes/Ccl5 influences hematopoietic stem cell subtypes and causes myeloid skewing
.
Blood
 
2012
;
119
:
2500
9
.

138.

Kusumbe
AP
,
Ramasamy
SK
,
Itkin
T
, et al.  
Corrigendum: age-dependent modulation of vascular niches for haematopoietic stem cells
.
Nature
 
2016
;
539
:
314
.

139.

Ferreira
MSV
,
Bergmann
C
,
Bodensiek
I
, et al.  
An engineered multicomponent bone marrow niche for the recapitulation of hematopoiesis at ectopic transplantation sites
.
J Hematol Oncol
 
2016
;
9
.

140.

Bruns
I
,
Lucas
D
,
Pinho
S
, et al.  
Megakaryocytes regulate hematopoietic stem cell quiescence through CXCL4 secretion
.
Nat Med
 
2014
;
20
:
1315
20
.

141.

Li
DT
,
Xue
WZ
,
Li
M
, et al.  
VCAM-1 macrophages guide the homing of HSPCs to a vascular niche
.
Nature
 
2018
;
564
:
119
24
.

142.

Chow
A
,
Lucas
D
,
Hidalgo
A
, et al.  
Bone marrow CD169 macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche
.
Journal of Experimental Medicine
 
2011
;
208
:
261
71
.

143.

Lee-Thedieck
C
,
Schertl
P
,
Klein
G
.
The extracellular matrix of hematopoietic stem cell niches q
.
Adv Drug Deliv Rev
 
2022
;
181
.

144.

Young
K
,
Eudy
E
,
Bell
R
, et al.  
Decline in IGF1 in the bone marrow microenvironment initiates hematopoietic stem cell aging
.
Cell Stem Cell
 
2021
;
28
:
1473
1482.e7
.

145.

Zhang
XY
,
Cao
DD
,
Xu
LT
, et al.  
Harnessing matrix stiffness to engineer a bone marrow niche for hematopoietic stem cell rejuvenation
.
Cell Stem Cell
 
2023
;
30
:
378
395.e8
.

146.

Ding
L
,
Saunders
TL
,
Enikolopov
G
,
Morrison
SJ
.
Endothelial and perivascular cells maintain haematopoietic stem cells
.
Nature
 
2012
;
481
:
457
62
.

147.

Acar
M
,
Kocherlakota
KS
,
Murphy
MM
, et al.  
Deep imaging of bone marrow shows non-dividing stem cells are mainly perisinusoidal
.
Nature
 
2015
;
526
:
126
30
.

148.

Hennrich
ML
,
Romanov
N
,
Horn
P
, et al.  
Cell-specific proteome analyses of human bone marrow reveal molecular features of age-dependent functional decline, nature
.
Communications
 
2018
;
9
.

149.

Zhou
R
,
Yang
GX
,
Zhang
Y
,
Wang
Y
.
Spatial transcriptomics in development and disease, molecular
.
Biomedicine
 
2023
;
4
:
4
.

150.

Lohoff
T
,
Ghazanfar
S
,
Missarova
A
, et al.  
Integration of spatial and single-cell transcriptomic data elucidates mouse organogenesis
.
Nat Biotechnol
 
2022
;
40
:
74
85
.

151.

Zhang
M
,
Eichhorn
SW
,
Zingg
B
, et al.  
Spatially resolved cell atlas of the mouse primary motor cortex by MERFISH
.
Nature
 
2021
;
598
:
137
43
.

152.

Zhang
D
,
Deng
YX
,
Kukanja
P
, et al.  
Spatial epigenome-transcriptome co-profiling of mammalian tissues
.
Nature
 
2023
;
616
:
113
22
.

153.

Ali
HR
,
Jackson
HW
,
Zanotelli
VRT
, et al.  
Imaging mass cytometry and multiplatform genomics define the phenogenomic landscape of breast cancer
.
Nat Cancer
 
2020
;
1
:
163
75
.

154.

Jackson
HW
,
Fischer
JR
,
Zanotelli
VRT
, et al.  
The single-cell pathology landscape of breast cancer
.
Nature
 
2020
;
578
:
615
20
.

155.

Berglund
E
,
Maaskola
J
,
Schultz
N
, et al.  
Spatial maps of prostate cancer transcriptomes reveal an unexplored landscape of heterogeneity
.
Nat Commun
 
2018
;
9
:
2419
.

156.

Ahmed
R
,
Zaman
T
,
Chowdhury
F
, et al.  
Single-cell RNA sequencing with spatial Transcriptomics of cancer tissues
.
Int J Mol Sci
 
2022
;
23
:
23
.

157.

Chen
WT
,
Lu
A
,
Craessaerts
K
, et al.  
Spatial Transcriptomics and In situ sequencing to study Alzheimer's disease
.
Cell
 
2020
;
182
:
976
991 e919
.

158.

Wang
M
,
Song
WM
,
Ming
C
, et al.  
Guidelines for bioinformatics of single-cell sequencing data analysis in Alzheimer's disease: review, recommendation, implementation and application
.
Molecular Neurodegeneration
 
2022
;
17
:
17
.

159.

Bressan
D
,
Battistoni
G
,
Hannon
GJ
.
The dawn of spatial omics
.
Science
 
2023
;
381
:
eabq4964
.

160.

Li
L
,
Bowling
S
,
McGeary
SE
, et al.  
A mouse model with high clonal barcode diversity for joint lineage, transcriptomic, and epigenomic profiling in single cells
.
Cell
 
2023
;
186
:
5183
5199.e22
.

Author notes

Xinrong Jin contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)