Abstract
Single-cell technologies have revealed the extensive heterogeneity and complexity of the immune system. Systems biology approaches in immunology have taken advantage of the high-parameter, high-throughput data and analyzed immune cell types in a ‘bottom-up’ data-driven method. This approach has discovered previously unrecognized cell types and functions. Especially for human immunology, in which experimental manipulations are challenging, systems approach has become a successful means to investigate physiologically relevant contexts. This review focuses on the recent findings in lymphocyte biology, from their development, differentiation into subsets, and heterogeneity in their functions, enabled by these systems approaches. Furthermore, we review examples of the application of findings from systems approach studies and discuss how now to leave the rich dataset in the curse of high-dimensionality.
Keywords: Plant-based, sustainability, Behaviour change, Social, Transition, Tipping points
Introduction
The immune system is comprised of diverse sets of cells with distinct yet versatile functions that interact with each other to exert sophisticated immune responses. For example, the powerful effect of vaccines we are witnessing against COVID is mediated by B cells which secrete antigen-specific antibodies, but the effective production of antibody production requires activated CD4+ helper T cells. CD4 T helper cells, on the other hand, require antigen presenting cells, such as dendritic cells. Depending on how they were stimulated, CD4 T cells differentiate into different subsets with distinct cytokine profiles, which shape various immune responses. What we call an immune response at an organismal level is actually this intricate coordination of all these diverse cell types. Thus, to comprehend physiologically relevant immune responses, it is critical to understand the diversity of immune cells in the act and their interactions with each other. Systems immunology is the attempt to understand the immune system as a whole and has successfully broadened the spectrum of immunological understanding1. One type of the most popular tools in systems immunology is single-cell analysis, the earliest form of which was flow cytometry. In the hematopoietic immune systems, cells are conventionally defined by their immunophenotype (i.e., CD3 as a T cell marker, CD19 as a B cell marker). At the same time, there are functional states assigned via their effector function (i.e., antigen presentation, phagocytosis, cytokine production). To capture both the diversity of cell types and states in a complex immune system, there is an inextricable link between single-cell techniques and the biological insights they can enable. Here, we review recent findings on lymphocyte differentiation, subsets, and functions by these systems approaches with single-cell technologies (Figure 1).
Figure 1.
Summary of lymphoid subsets and functions reviewed in this article. This article reviews the recent systems approach studies on lymphopoiesis in bone marrow and thymus and cell subtypes and functions of innate lymphoid cell, B cell, and T cell.
Systems Analysis with Single-cell technologies
The most familiar and established single-cell technique immunologists have is the fluorescence-activated cell sorter (FACS)2. The concept of labeling cells with fluorescent-tagged monoclonal antibodies and then analyzing them one at a time in the flow cytometer was first described by Leonard Herzenberg in 19692,3. Furthermore, the prospective isolation (or “sorting”) functionality of FACS enable downstream functional experiments with the exact cells identified by their flow cytometric properties. Ever since then, immunologists have identified, prospectively isolated, and analyzed the functions of immune cell types that match with a specific immune phenotype or master regulator transcription factor (i.e. CD25+ FoxP3+ regulatory T cells). Considering the widespread usage of this powerful tool in the history of immunology research, it is not an overstatement to say that modern immunology is based on the flow cytometry. Still, the limitation in the parameterization of these experiments (i.e. multiplexing) meant that most applications were still ‘top-down’, where cell populations of interest and their compositions were pre-determined prior to the experiment.
Single-cell mass cytometry, also known as Cytometry by Time-of-flight (CyTOF), is a direct successor of flow cytometry with metal-isotope-tagged antibodies4,5, instead of fluorescence-tagged antibodies. By measuring the metal-isotope-specific mass-to-charge ratio (m/z), mass cytometry increases the number of parameters measured by the cell to over 50 per cell on millions of cells per experiment5,6. Inheriting many aspects of flow cytometry (sample processing, staining, similar machine operations, data structure format, etc.), the increased number of parameters enabled a smooth transition of flow cytometry users into a systems approach with this tool. The technical advantages of mass cytometry and comparison with flow cytometry have been described well in other reviews6,7. Overall, this increased parameterization, facilitates a more ‘bottom-up’, data-driven understanding of each experiment where previously unanticipated cell states and populations could be discovered and organized through unanticipated combinations of the molecular features measured.
With higher parameterization, but also higher cost-per-cell and therefore relatively lower cell number, single-cell sequencing has become more common and accessible to all fields of biology. Ever since its first appearance in 20098, we have experienced a burst of different methods and throughput in single-cell RNA-sequencing (scRNA-seq) in the last decade9. The recent publication of Tabula Sapiens10 with nearly 500,000 cells from 24 tissues and organs, and similarly Tabula Muris a few years earlier11, highlights the widespread application of scRNA-seq in different tissues as well as the abundance of public resources that can be utilized in a systems approach. Following the widespread impact of scRNA-seq in immunology research12–14, technologies to analyze other modalities, such as chromatin accessibility15,16, histone modifications17, chromatin conformation18, etc., have also become available as high-throughput single-cell assays. More recently, new methods that combine measurements of multiple modalities, including RNA, protein epitopes, chromatin accessibility, spatial information, and more, have developed rapidly. Multi-modal technologies and spatial information provide novel opportunities to study complex regulatory mechanisms and cell-cell interactions, as reviewed elsewhere19,20. For the scope of this article, we will focus on the biological findings on lymphocytes made with CyTOF and single-cell sequencing methods.
Single-cell techniques highlight the continuum of adult human hematopoiesis
The bottom-up, data-driven interpretation of next-generation single-cell datasets, has enabled researchers to investigate systems that are traditionally difficult to experimentally manipulate, such as human hematopoiesis bone marrow cells. Such approaches now allow the construction of dynamic models of cellular differentiation processes from an otherwise static sample because virtually all cell types and states are present at any one time and can be organized in silico. While the concept of hematopoiesis and hematopoietic stem and progenitors have been defined both in mice and humans, human bone marrow is not nearly accessible to intervene or experiment in its native condition compared to mouse models. Hence, with the advances in single-cell technologies, multiple groups have investigated human bone marrow hematopoiesis and demonstrated the molecular phenotypic continuum of hematopoietic progenitors in protein5, RNA21,22, and chromatin accessibility23 landscapes. Such collection of high-throughput and high-dimensional data that span the whole developmental trajectory provided opportunities for the development of trajectory inference (TI) algorithms – often referred to as pseudotime analysis. In these computational techniques, cells are ordered by similarity in high dimensional (RNA, protein, chromatin accessibility, etc.) space, with the aim of recapitulating the natural differentiation process in vivo24. Among multiple TI algorithms developed recently24,25, one of the early exemplary uses of TI was a study with B lymphopoiesis in human bone marrow26. In this study, the authors developed a TI algorithm, Wanderlust, to re-create the B lymphopoiesis pseudotime based on a single cell mass cytometry analysis of bone marrow progenitors. Across pseudotime, there were specific coordination points at which multiple developmentally crucial events, such as the IL-7/STAT5 checkpoint which leads to the immunoglobulin gene rearrangement event. Moreover, the developmental trajectory of human B lymphopoiesis established in this paper became the basis for analyzing disease samples in another study with B cell precursor acute lymphoblastic leukemia27. Good et al. took a machine learning approach to assign B-leukemic blasts to the closest normal B-cell developmental population and identified developmentally dependent predictors of relapse that significantly enhance the risk prediction at diagnosis in acute lymphoblastic leukemia (ALL)27. Similar approaches of utilizing normal single-cell hematopoietic data to analyze malignant leukemic samples have been used with RNA expression or chromatin accessibility data as well28,29.
Along with the single-cell RNA-seq and trajectory inferences, novel methods for lineage tracing have evolved rapidly over the last decade30. Especially the combination of single-cell RNA-seq as a readout of lineage barcodes empowered scientists to compare the ground truth trajectory by the lineage barcode to the trajectory inferred from the transcriptome of single cells31. Applying this method in hematopoiesis, Weinreb et al. revealed cell fate biases are made clonally early in differentiation, and also that clonally distinct hematopoietic progenitors can converge into a similar transcriptome, such as the monocyte development in the mouse steady-state hematopoiesis32. These methods and findings signify how the systems approaches using single-cell methods are evolving to expand our knowledge.
T lymphopoiesis in human thymus examined by advanced tools
Another human hematopoietic organ that has been largely inaccessible for experimental manipulation is the thymus, in which later T cell development takes place. While the complex steps of thymopoiesis have been studied extensively owing to mouse models33, the translation of paradigms to human systems has been challenging, in part due to the scarcity of human thymus samples. Thus, recent scRNA-seq studies of human thymocytes34–36 have considerably filled the knowledge gap in the field. By assaying single-cell transcriptome from more than 250,000 pre-natal and post-natal thymic cells, Park et al. established a framework of human thymus development. Among 50 different cell states identified in the human thymus, including fibroblasts and epithelial cells, existed unconventional T cells that were first reported in human thymus, such as CD8αα+ T cells, NKT-like cells, and TH17-like cells34. Another approach used by Le et al. and Lavaert et al. was to enrich the most premature precursors, earliest thymic progenitors (ETPs), by enriching for CD34+ cells in the thymus via FACS to study T-lineage developmental progression. Both groups utilized high-dimensional scRNA-seq data to define the developmental states of thymocytes and showed that the overall trajectory and expression of core regulatory genes are conserved between mouse and human thymopoiesis35–37. In particular, both studies identified a progenitor population expressing the IL-3 receptor subunit CD123, that is transcriptionally primed for plasmacytoid dendritic cells (pDCs) in the thymus CD34+ compartment35,36. One remaining question is how these putative ETPs are connected to other cells in the hematopoietic hierarchy. While the fetal liver lympho-myeloid progenitors38 seemed to bear the closest transcriptomic profile compared to the fetal thymic ETPs34, the bone marrow counterpart remains ambiguous.
T cell heterogeneities explained at the single-cell level
Even for T cells, likely the most studied cell type in immunology, the heterogeneity across different tissue sites and activation states has been a difficult topic to tackle. For example, T cell exhaustion has been described for decades but the term has been used broadly for a heterogeneous population of exhausted or dysfunctional T cells39–41. Alongside the seminal papers in the field that identified TOX as the exhaustion marker via more traditional methods42,43, numerous studies utilized scRNA-seq to analyze the heterogeneity of T cell responses and discovered key gene regulation programs led by TOX44 and TCF-145, driving these cell states. Especially the expression of TOX and the epigenetic landscaped driven by TOX is demonstrated to be the crucial difference between other memory and effector T cells versus exhausted T cells42–44,46,47. The study of exhausted T cells using similar approaches has been extended to the context of chronic infection, cancer, and cancer immunotherapy with single-cell technologies48–51 whose findings can be reviewed elsewhere40,41. Moreover, the VDJ sequencing of TCRs has enabled clonal lineage tracing of T cells in single-cell sequencing approaches. In the context of T cell exhaustion, TCR sequencing was successfully applied in the basal cell carcinoma samples with checkpoint blockade to show that novel clonotypes of T cell expansion upon anti-PD-1 treatment52. Another recent study with TCR sequencing showed that TCR signaling avidity correlated with different subsets of exhausted T cells in mouse LCMV models or human tumor-infiltrating lymphocytes53.
Another crucial aspect of T cells that is frequently associated with their functional states is the cellular metabolism54,55. While the significance of metabolic regulation for different T cell populations has been recognized previously56,57, the bottom-up approaches to utilize the metabolic states of T cells to investigate their functional states at the single-cell level is fairly new. Recently, protein-based single-cell studies that specifically targeted the metabolic wiring of CD8 T cells reported metabolic states in accordance with functional states at the single-cell level58,59. These two studies utilized mass cytometry to quantify the protein expressions of regulators that served as surrogates to the overall activity of the respective pathway they resided in58. Specifically, Hartman et al. demonstrated that, more precisely than the broader expression of exhausted T cell phenotypes CD39 and PD1, the metabolic state indicators specifically corroborate the exhausted cell states, particularly within the tumor microenvironment58. Further discussions on T cell immunometabolism can be found in other review articles60,61.
To reinforce recent advances, researchers have proposed an updated T cell atlas. Szabo et al., for instance, have described the tissue-specific signatures and CD4 or CD8 cell-type-specific activation states over 50,000 human T cells based on their transcriptomic profiles62. Orthogonally, Wang et al. have compared classical T cell subsets by surface markers to T cell populations defined by scRNA-seq profiles63. While these studies suggest more complex T cell subtypes than conventionally defined, the authors conducted comparison with sorted populations to bridge the gap with what T cell biologists currently use and what is observed in single-cell analyses. Moreover, the promise of data-driven bottom-up analysis of complex single cell datasets is on full display where we are now able to differentiate all cellular T cell diversity within an immune tissue in one analytical pass.
B cell classification revisited with a systems approach
Compared to a plethora of T cell studies with systems approaches, there have been limited attempts to systematically investigate the functional states of human B cells. In most cases, B cell classification has been based on their maturation status, antibody repertoire, and antigen-experience states64. As an example, single-cell sequencing combined with VDJ sequencing was utilized to study the B cell maturation trajectory during antibody class switching in tonsils65. Although antibody production is exclusively a B-cell-specific function, the other functions of B cells, such as antigen presentation, and cytokine production, as well as tissue-specific B cell subsets should not be neglected. In this regard, a recent mass cytometry study screened for the expression of 351 surface molecules on human B cells in 4 lymphoid tissues (bone marrow, peripheral blood, tonsil, and lymph node)66. In this, Glass et al. identified 12 different populations of human B cells and interrogated their functions via subset-specific protein expression profiles. Among the 12 populations exist a CD19hiCD11c+ memory population that exhibited active metabolic and transcriptional state and a CD39+ tonsil-resident population66. Moreover, the authors defined the gating schemes, CD45RB to the definition of early B cell memory, for populations identified from their study.66 Studies like this, that connect high dimensional multi-omic datasets to ‘human-interpretable’ cell populations in low dimensional space, provide a valuable resource for more focused studies to come.
Innate Lymphoid Cells: Newest members of the family
Over the last decade and a half, immunologists continue to discover new NK-like lymphoid cell types (i.e., non-T and -B) that exhibit distinct cytokine production profiles67,68. They were soon given a uniform nomenclature as different groups of innate lymphoid cells (ILCs) based on the absence of rearranged antigen receptors69 and classified together with already well-known NK cells and lymphoid tissue inducer (LTi), due to their phenotypic similarities and common developmental progenitors67. As their critical roles in tissue homeostasis are emphasized, recent ILC studies highlight the tissue-specific transcriptomic programs which suggest tissue microenvironment-derived priming during ILC differentiation70–73.
On the other hand, NK cells, the quintessential ILC, have historically been characterized as circulating cells in peripheral blood. Nonetheless, NK cells in peripheral blood already show a vast diversity of NK receptor expression phenotypes, as demonstrated in the single-cell mass cytometry study by Horowitz et al.. In this study, the authors concluded that the combinatorial expression of NK receptors could lead to an estimated 30,000 NK cell phenotypic populations in an individual74. Subsequently, various groups have utilized scRNA-seq to further describe NK cell diversity in mice and humans75–77, in which tissue-specific NK subsets and differentiation trajectories were suggested. Considering their ability as a front-line defense against viruses and cancer, we anticipate a deeper understanding of NK cells to benefit the emerging NK cell-based immunotherapies78,79.
Discussions – Implications of Immune Heterogeneity
Single-cell studies have highlighted the immense diversity of cell types within the immune system where previously thought of as discrete cell populations contain new molecular phenotypes that are often continuously connected to one another. Still, systems approach studies rely on computational clustering algorithms to define populations, whose granularity can be set manually by a parameter. Hence, the old, but still valid, debate between “lumpers and splitters” is back. How granular should one be in defining a cell type? Where is the bar between the plasticity of an immune cell and a distinct cell type? Single-cell studies without specific biological questions or functionally assayable endpoints could end up with broad but shallow analyses lacking meaningful insights.
To provide worthwhile knowledge to the field, a systems approach should be incorporated into the research as a means, not a goal. It is fundamental that studies on cell types and functional subsets need to be linked to specific definitions such that a ‘novel’ population can be the subject of follow-up studies. In any given modality, either a cell-type-specific phenotype or cell-type-specific signature matrix should be defined to minimize ambiguity and/or subjectivity and enable more mechanistic studies and validation of causal relationships (Figure 2). Researchers should also consider the implication of a population in healthy and diseased context before claiming a new cell type to understand its role in homeostasis versus response to disruption.
Figure 2.
Example of high-dimensional data from a systems study translated into low-dimensional space and applied for follow-up studies. (Top) High-dimensional data from a systems approach is often analyzed with dimensional reduction algorithms or heatmaps. Based on this rich dataset, researchers often find a target population of interest (marked by red boxes). It is critical to provide some type of signature phenotype for populations. (Middle) With the signature phenotype, the target population is now identifiable in the low-dimension analysis and can be prospectively isolated. (Bottom) Then, this population can be re-analyzed in the high-dimensional data to validate the cell identity and subjected to follow-up experiments and applications.
Albeit complicated, a well-designed, purpose-driven systems approach can provide the most profound and transferable information in multiple layers. It is undeniable that the burst of new technological advances and the systems approach studies utilizing them are contributing to understanding the immune system more rapidly than ever.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Davis MM, Tato CM & Furman D Systems immunology: Just getting started. Nature Immunology vol. 18 725–732 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bonner WA, Hulett HR, Sweet RG & Herzenberg LA Fluorescence activated cell sorting. Rev. Sci. Instrum 43, 404–409 (1972). [DOI] [PubMed] [Google Scholar]
- 3.Hulett HR, Bonner WA, Barrett J & Herzenberg LA Cell sorting: Automated separation of mammalian cells as a function of intracellular fluorescence. J. Immunol 193, 2045–2047 (2014). [PubMed] [Google Scholar]
- 4.Bandura DR et al. Mass Cytometry : Technique for Real Time Single Cell Multitarget Immunoassay Based on Inductively Coupled Plasma Time-of-Flight Mass Spectrometry. Anal. Chem 81, 6813–6822 (2016). [DOI] [PubMed] [Google Scholar]
- 5.Bendall SC et al. Single-Cell Mass Cytometry of Differential Immune and Drug Responses Across a Human Hematopoietic Continuum. Science (80-.) 332, 687–695 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bjorson ZB, Nolan GP & Fantl WJ Single Cell Mass Cytometry for Analysis of Immune System Functional States. Curr. Opin. Immunol 25, (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bendall SC, Nolan GP, Roederer M & Chattopadhyay PK A deep profiler’s guide to cytometry. Trends Immunol. 33, 323–332 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tang F et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009). [DOI] [PubMed] [Google Scholar]
- 9.Aldridge S & Teichmann SA Single cell transcriptomics comes of age. Nature Communications vol. 11 1–4 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jones RC et al. The Tabula Sapiens: A multiple-organ, single-cell transcriptomic atlas of humans. Science (80-.) 376, (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Schaum N et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Stubbington MJT, Rozenblatt-Rosen O, Regev A & Teichmann SA Single-cell transcriptomics to explore the immune system in health and disease. Science vol. 358 58–63 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Papalexi E & Satija R Single-cell RNA sequencing to explore immune cell heterogeneity. Nature Reviews Immunology vol. 18 35–45 (2018). [DOI] [PubMed] [Google Scholar]
- 14.Yost KE, Chang HY & Satpathy AT Tracking the immune response with single-cell genomics. Vaccine 38, 4487–4490 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.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 10, 1213–1218 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cusanovich DA et al. Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing. Science (80-.) 348, 910–914 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kaya-Okur HS et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun 10, 1–10 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ramani V et al. Massively multiplex single-cell Hi-C. Nat. Methods 14, 263–266 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhu C, Preissl S & Ren B Single-cell multimodal omics: the power of many. Nature Methods vol. 17 11–14 (2020). [DOI] [PubMed] [Google Scholar]
- 20.Ma A, McDermaid A, Xu J, Chang Y & Ma Q Integrative Methods and Practical Challenges for Single-Cell Multi-omics. Trends in Biotechnology vol. 38 1007–1022 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Velten L et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol 19, 271–281 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pellin D et al. A comprehensive single cell transcriptional landscape of human hematopoietic progenitors. Nat. Commun 10, 2395 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Buenrostro JD et al. Integrated Single-Cell Analysis Maps the Continuous Regulatory Landscape of Human Hematopoietic Differentiation. Cell 173, 1535–1548.e16 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Saelens W, Cannoodt R, Todorov H & Saeys Y A comparison of single-cell trajectory inference methods. Nat. Biotechnol 37, 547–554 (2019). [DOI] [PubMed] [Google Scholar]
- 25.Deconinck L, Cannoodt R, Saelens W, Deplancke B & Saeys Y Recent advances in trajectory inference from single-cell omics data. Current Opinion in Systems Biology vol. 27 100344 (2021). [Google Scholar]
- 26.Bendall SC et al. Single-cell trajectory detection uncovers progression and regulatory coordination in human b cell development. Cell 157, 714–725 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Using high-dimensional proteomic landscapes measured via CyTOF, the authors constructed pseudotime of B cell development in human bone marrow and defined key developmental checkpoints, such as IL-7/STAT5 signaling, which could be experimentally interrupted to inhibit B cell development.
- 27.Good Z et al. Single-cell developmental classification of B cell precursor acute lymphoblastic leukemia at diagnosis reveals predictors of relapse. Nat. Med 24, 474–483 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]; * Based on the B cell developmental trajectory by Bendall et al. (2014), the authors demonstrated which developmental stages BCP-ALL cells are and successfully identified cues of relapse based on the developmental-stage specific features.
- 28.Corces MR et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet 48, 1193–1203 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Granja JM et al. Single-cell multiomic analysis identifies regulatory programs in mixed-phenotype acute leukemia. Nat. Biotechnol 37, 1458–1465 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.VanHorn S & Morris SA Next-Generation Lineage Tracing and Fate Mapping to Interrogate Development. Developmental Cell vol. 56 7–21 (2021). [DOI] [PubMed] [Google Scholar]
- 31.Wagner DE & Klein AM Lineage tracing meets single-cell omics: opportunities and challenges. Nat. Rev. Genet (2020) doi: 10.1038/s41576-020-0223-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Weinreb C, Rodriguez-Fraticelli A, Camargo FD & Klein AM Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science (80-.) 367, eaaw3381 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Coupling lineage tracing with single-cell RNA-seq on in vivo and in vitro mouse hematopoietic stem and progenitor cells, Weinreb and Rodriguez-Fraticelli dissected the fate decisions made in clonal levels.
- 33.Yui MA & Rothenberg EV Developmental gene networks: A triathlon on the course to T cell identity. Nat. Rev. Immunol 14, 529–545 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Park J-E et al. A cell atlas of human thymic development defines T cell repertoire formation. Science (80-.) 367, eaay3224 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Park et al. provided the first single-cell atlas of human thymus and identified multiple novel cell types in primary human thymus samples.
- 35.Lavaert M et al. Integrated scRNA-Seq Identifies Human Postnatal Thymus Seeding Progenitors and Regulatory Dynamics of Differentiating Immature Thymocytes. Immunity 52, 1088–1104.e6 (2020). * [DOI] [PubMed] [Google Scholar]
- 36.Le J et al. Single-Cell RNA-Seq Mapping of Human Thymopoiesis Reveals Lineage Specification Trajectories and a Commitment Spectrum in T Cell Development. Immunity 52, 1105–1118.e9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; * Lavaert et al. and Le et al. describe the earliest thymus progenitors and their differentiation pathway into T cells or other cell types in human thymus.
- 37.Rothenberg EV Single-cell insights into the hematopoietic generation of T-lymphocyte precursors in mouse and human. Exp. Hematol (2021) doi: 10.1016/j.exphem.2020.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Popescu D-M et al. Decoding human fetal liver haematopoiesis. Nature (2019). doi: 10.1038/s41586-019-1652-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wherry EJ T cell exhaustion. Nature Immunology vol. 12 492–499 (2011). [DOI] [PubMed] [Google Scholar]
- 40.McLane LM, Abdel-Hakeem MS & Wherry EJ CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. Annual Review of Immunology vol. 37 457–495 (2019). [DOI] [PubMed] [Google Scholar]
- 41.Blank CU et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol 19, 665–674 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]; * Through a thorough discussion on what T cell exhaustion is, the authors summarize recent findings in T cell exahustion and the challenges and diretions of the field.
- 42.Khan O et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Alfei F et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019). [DOI] [PubMed] [Google Scholar]
- 44.Yao C et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol 20, 890–901 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chen Z et al. TCF-1-Centered Transcriptional Network Drives an Effector versus Exhausted CD8 T Cell-Fate Decision. Immunity 51, 840–855.e5 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Scott AC et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Seo H et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion. Proc. Natl. Acad. Sci. U. S. A 116, 12410–12415 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kurtulus S et al. Checkpoint Blockade Immunotherapy Induces Dynamic Changes in PD-1 − CD8 + Tumor-Infiltrating T Cells. Immunity 50, 181–194.e6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sade-Feldman M et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 176, 404 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Miller BC et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol 20, 326–336 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zander R et al. CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer. Immunity 51, 1028–1042.e4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yost KE et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med 25, 1251–1259 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Daniel B et al. Divergent clonal differentiation trajectories of T cell exhaustion. Nat. Immunol 23, 1614–1627 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Buck MD, Sowell RT, Kaech SM & Pearce EL Metabolic Instruction of Immunity. Cell vol. 169 570–586 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Geltink RIK, Kyle RL & Pearce EL Unraveling the Complex Interplay between T Cell Metabolism and Function. Annual Review of Immunology vol. 36 461–488 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Maciver NJ, Michalek RD & Rathmell JC Metabolic regulation of T lymphocytes. Annual Review of Immunology vol. 31 259–283 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.MacIver NJ & Rathmell JC Editorial overview: Metabolism of T cells: integrating nutrients, signals, and cell fate. Current Opinion in Immunology vol. 46 viii–xi (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hartmann FJ et al. Single-cell metabolic profiling of human cytotoxic T cells. Nat. Biotechnol 39, 186–197 (2021). * [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Levine LS et al. Single-cell analysis by mass cytometry reveals metabolic states of early-activated CD8+ T cells during the primary immune response. Immunity 54, 829–844.e5 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]; * Hartmann et al. and Levine et al. demonstrate mass cytometry assay to measure enzymes in metabolic pathways that succesfully recaptiulate the metabolic state of the cell, enabling measurement of metabolic state of single-cells.
- 60.Artyomov MN & Van den Bossche J Immunometabolism in the Single-Cell Era. Cell Metabolism vol. 32 710–725 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Purohit V, Wagner A, Yosef N & Kuchroo VK Systems-based approaches to study immunometabolism. Cellular and Molecular Immunology vol. 19 409–420 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Szabo PA et al. Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease. Nat. Commun 10, 1–16 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang X et al. Reinvestigation of Classic T Cell Subsets and Identification of Novel Cell Subpopulations by Single-Cell RNA Sequencing. J. Immunol 208, 396–406 (2022). [DOI] [PubMed] [Google Scholar]
- 64.Maecker HT, McCoy JP & Nussenblatt R Standardizing immunophenotyping for the Human Immunology Project. Nature Reviews Immunology vol. 12 191–200 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.King HW et al. Single-cell analysis of human B cell maturation predicts how antibody class switching shapes selection dynamics. Sci. Immunol 6, 6291 (2021). [DOI] [PubMed] [Google Scholar]
- 66.Glass DR et al. An Integrated Multi-omic Single-Cell Atlas of Human B Cell Identity. Immunity 53, 217–232.e5 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; * Comprehensive immunophenotypic survey of human B cell identity across tissues. Measuring 351 surface molecule expression patterns on human B cells from 4 different tissues, Glass et al. reconcile tissue-specifc molecular and phenotypic differences in B cell populations and lay out role for CD45RB in human B cell memory.
- 67.Vivier E et al. Innate Lymphoid Cells: 10 Years On. Cell vol. 174 1054–1066 (2018). [DOI] [PubMed] [Google Scholar]
- 68.Bal SM, Golebski K & Spits H Plasticity of innate lymphoid cell subsets. Nature Reviews Immunology vol. 20 552–565 (2020). [DOI] [PubMed] [Google Scholar]
- 69.Spits H et al. Innate lymphoid cells-a proposal for uniform nomenclature. Nat. Rev. Immunol 13, 145–149 (2013). [DOI] [PubMed] [Google Scholar]
- 70.Yudanin NA et al. Spatial and Temporal Mapping of Human Innate Lymphoid Cells Reveals Elements of Tissue Specificity. Immunity 50, 505–519.e4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Meininger I et al. Tissue-Specific Features of Innate Lymphoid Cells. Trends in Immunology vol. 41 902–917 (2020). [DOI] [PubMed] [Google Scholar]
- 72.Mazzurana L et al. Tissue-specific transcriptional imprinting and heterogeneity in human innate lymphoid cells revealed by full-length single-cell RNA-sequencing. Cell Res. 31, 554–568 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]; * Summarizing recent finding in ILC studies, the authors summarize the phenotypes of ILCs across tissues in both humans and mice
- 73.McFarland AP et al. Multi-tissue single-cell analysis deconstructs the complex programs of mouse natural killer and type 1 innate lymphoid cells in tissues and circulation. Immunity 54, 1320–1337.e4 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Horowitz A et al. Genetic and environmental determinants of human NK cell diversity revealed by mass cytometry. Sci. Transl. Med 5, (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Crinier A et al. High-Dimensional Single-Cell Analysis Identifies Organ-Specific Signatures and Conserved NK Cell Subsets in Humans and Mice. Immunity 49, 971–986.e5 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Yang C et al. Heterogeneity of human bone marrow and blood natural killer cells defined by single-cell transcriptome. Nat. Commun 10, 1–16 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Smith SL et al. Diversity of peripheral blood human NK cells identified by single-cell RNA sequencing. Blood Adv. 4, 1388–1406 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wu SY, Fu T, Jiang YZ & Shao ZM Natural killer cells in cancer biology and therapy. Molecular Cancer vol. 19 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Albinger N, Hartmann J & Ullrich E Current status and perspective of CAR-T and CAR-NK cell therapy trials in Germany. Gene Therapy vol. 28 513–527 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]