Structured Abstract
Purpose of review:
Myeloid cells – granulocytes, monocytes, macrophages and dendritic cells (DCs) – are innate immune cells that play key roles in pathogen defense and inflammation, as well as in tissue homeostasis and repair. Over the past 5 years, in part due to more widespread use of single cell omics technologies, it has become evident that these cell types are significantly more heterogeneous than was previously appreciated. In this review we consider recent studies that have demonstrated heterogeneity among neutrophils, monocytes, macrophages and DCs in mice and humans. We also discuss studies that have revealed the sources of their heterogeneity.
Recent findings:
Recent studies have confirmed that ontogeny is a key determinant of diversity, with specific subsets of myeloid cells arising from distinct progenitors. However, diverse microenvironmental cues also strongly influence myeloid fate and function. Accumulating evidence therefore suggests that a combination of these mechanisms underlies myeloid cell diversity.
Summary:
Consideration of the heterogeneity of myeloid cells is critical for understanding their diverse activities, such as the role of macrophages in tissue damage versus repair, or tumor growth versus elimination. Insights into these mechanisms are informing the design of novel therapeutic approaches.
Keywords: Myeloid cells, Heterogeneity, Myelopoiesis, Progenitors, Microenvironment
Introduction
Granulocytes, monocytes, macrophages and dendritic cells (DCs) are collectively known as myeloid cells. Most are produced by hematopoietic stem cells (HSCs), although some arise early in development prior to the appearance of HSCs. In recent years, studies employing single-cell transcriptomic (scRNAseq) and multiparametric protein (flow cytometry and CyTOF) profiling have revealed notable heterogeneity within the myeloid lineages. In this review, we will consider how heterogeneity arises when myeloid cells originate from distinct tissues (e.g. yolk sac, fetal liver, bone marrow, or spleen) or are produced at different times (e.g. during fetal development, the neonatal period, or adulthood), and how microenvironmental cues also underlie myeloid subset diversity between organs and even within them (illustrated in Figure 1). We will also consider the functional relevance of myeloid cell heterogeneity.
Figure 1. Sources of myeloid cell heterogeneity.

(A) Sites of myelopoiesis. Early in development, prior to the emergence of hematopoietic stem cells (HSCs), macrophages are produced by erythromyeloid progenitors (EMPs) in the yolk sac and populate the developing embryonic tissues. Later, HSCs arising in the AGM region and stored in the fetal liver produce monocytes, neutrophils and DCs, and monocyte-derived macrophages are supplied to tissues and organs. After birth, HSCs are stored in the bone marrow and continue to produce myeloid cells throughout life. Myelopoiesis can also occur in the spleen, especially under inflammatory conditions, and to a lesser degree in inflamed tissues. In adults, tissue-resident macrophages have diverse origins – some remain yolk sac-derived, others are fetal monocyte-derived, and some are constantly replaced by newly recruited monocytes. (B) Myeloid cell heterogeneity can reflect distinct progenitor origins in the bone marrow, premature release of immature cells (especially under inflammatory conditions), and the influence of diverse microenvironmental cues between and even within organs.
The Heterogeneity and Origins of Neutrophils and Other Granulocytes
Among the granulocytes, neutrophils are the best studied (1) and the most abundant cell type in the bone marrow, where they can be found at different stages of differentiation from hematopoietic progenitors. They are typically released into the circulation as mature neutrophils, but under inflammatory conditions immature neutrophils may be found in the circulation. At the end of their lifecycle, neutrophils return to the bone marrow.
Recent studies have demonstrated the sequential development of neutrophils from early to intermediate and late precursor cell stages with specific surface marker and transcriptomic signatures (Figure 2). An early committed neutrophil progenitor (proNeu1) expressing Ly6C and CD81 but lacking CD11b was recently characterized in the LKS− CD34+ FcγRhi fraction of mouse bone marrow (2), downstream of oligopotent Granulocyte-Monocyte Progenitors (GMPs), which produce both monocytes and neutrophils. Neutrophil development from proNeu1 then proceeds via proNeu2, which have begun to express CD11b and represent a transitional stage that gives rise to pre-neutrophils (preNeu) (2). preNeus have also been further subdivided into preNeu 1, 2 and 3, which are thought to represent hierarchical stages of neutrophil differentiation (3). preNeus subsequently differentiate into non-proliferative immature neutrophils (immNeu), which mature in the bone marrow to yield mature neutrophils (mNeus) (4). Human equivalents of proNeus and preNeus have also been described (2, 5).
Figure 2. Neutrophil differentiation and diversity.

Neutrophils are derived from granulocyte-monocyte progenitors (GMPs, which also yield monocytes) via neutrophil-committed proNeu1 progenitors, then proNeu2 cells that begin expressing CD11b, 3 stages of preNeus, immature neutrophils (immNeu) and finally mature neutrophils (mNeu). Blood neutrophils include PMNa (arise from immNeu and mNeu), PMNb (mNeu-derived), and PMNc (derived from both PMNa and PMNb).
Three subsets of mature neutrophils have recently been described in bone marrow and blood: PMNa, PMNb and PMNc (6). PMNa are enriched in S100a8, S100a9 and Mmp8 gene expression, whereas PMNb express interferon-stimulated genes. PMNc have characteristics of senescent cells and are enriched in genes for ribosome biogenesis, cytoplasmic translation, post-transcriptional regulation and LPS-mediated signaling pathways. In the steady-state, PMNa arise from both mNeu and immNeu, PMNb from mNeu, and PMNc from both PMNa and PMNb (Figure 2). In contrast, E. coli infection suppresses differentiation of immNeu to PMNa, and immNeu mainly differentiate into mNeu. Neutrophils can also originate from different bone marrow sites. For instance, skull and vertebral bone marrow niches have been reported to specifically supply neutrophils to the meninges (7**).
Phenotypically diverse neutrophils can be found in healthy tissues, where they can acquire tissue-specific phenotypes under the influence of tissue-derived signals (8), but neutrophil heterogeneity is even more evident under disease conditions, such as inflammation, infection or cancer, including subsets that promote tumor growth and others that are anti-tumorigenic (9–11). Interestingly, a live imaging study recently demonstrated variation in neutrophil shape and movement that permitted the definition of “behavioral landscapes” of neutrophil subsets during inflammation (12**).
Two subsets of neutrophils have been identified within the red pulp of the mouse spleen: an immature, immobile Ly6Gint population and a more differentiated and mobile Ly6Ghi population (13). Upon Streptococcus pneumoniae infection, Ly6Ghi neutrophils pull bacteria off the surface of red pulp macrophages to kill them, while Ly6Gint neutrophils proliferate to increase the number of mature neutrophils. During S. aureus infection, neutrophils with the ability to recruit additional neutrophils to fight the infection enter the lymph nodes via L-selectin (14).
Neutrophils with high expression of MHCII and the ability to activate CD4 T cells ex vivo have also been observed in uninfected lymph nodes (15). Moreover, during inflammation, neutrophils have been reported to transdifferentiate into cells with features of both neutrophils and dendritic cells, called PMN-DCs (16). They have higher expression of pattern recognition receptors and kill fungal cells more efficiently than canonical neutrophils, but they can also trigger an efficient adaptive response against fungi after antigen pulsing by inducing Th1 and Th17 responses.
To better understand heterogeneity among neutrophils, a single phenotypic spectrum ordered chronologically and named “neutrotime” has recently been suggested (17*). The authors propose that phenotypic divergence among neutrophils arises from deviation from a specific position within the neutrotime continuum due to the influence of microenvironmental cues, and that each neutrophil maintains features from the point of deviation in neutrotime and acquires features according to the sequence of signals encountered. This mechanism provides an opportunity to generate a highly diverse neutrophil repertoire without a requirement for committed developmental branches or cell subsets. Therefore, neutrophil heterogeneity could result from both intrinsic cell programming by transcription factors during differentiation and the influence of external signals.
There is also accumulating evidence that other granulocytes (eosinophils, basophils and mast cells) are similarly heterogeneous (18–20). For instance, like macrophages (discussed in more detail below), mast cells, which are also tissue-resident, have dual developmental origins from both yolk sac progenitors and fetal liver/bone marrow HSCs, and mast cells in different tissues express common as well as tissue-specific gene signatures (21).
The Heterogeneity of Monocytes and Macrophages
Macrophages are the earliest immune cells to emerge during development, with production originating in the yolk sac where they arise from erythromyeloid progenitors during primitive hematopoiesis and begin to populate the tissues of the developing embryo (Figure 1). Once definitive hematopoiesis is established in the embryo itself, monocytes are produced by HSCs, which are initially maintained in the fetal liver and later in the bone marrow. Some resident macrophages of yolk sac or fetal monocyte origin are rapidly or gradually replaced over time, whereas other persist for decades or even a whole lifetime. Thus, macrophage heterogeneity in tissues is, at least in part, a reflection of their developmental origins.
The function of macrophages can also be tissue-dependent (22, 23). Although they share many key features, macrophages in different tissues, and even within the same tissue, can have distinct gene expression profiles due to the influence of microenvironmental cues. Some microenvironmental signals are relatively stable, such as those that define tissue identity, whereas others may reflect responses to infection or injury. Recent studies have also shown that monocytes and macrophages retain “memory” of previous exposures to microbial and inflammatory stimuli, which persists due to epigenetic and metabolic changes (24). Thus, within a tissue, the current state of each macrophage reflects its ontogeny and tissue location, as well as its past and current experiences.
Additional insight into these concepts was provided by a recent analysis of mouse tissue-resident macrophages that revealed 3 broadly conserved subsets in diverse organs, including the heart, liver, lung, kidney, and brain, albeit with different relative abundances in each tissue (25**). Macrophages expressing TIMD4/LYVE1/FOLR2 (TLF+ macrophages) have embryonic and early fetal origins and are maintained by self-renewal, whereas CCR2+ macrophages are monocyte-derived and constantly replaced. The third subset lacks TIMD4, LYVE1, FOLR2 and CCR2, but expresses high levels of MHCII and appears to be more slowly and only partially maintained by monocyte recruitment. Interestingly, TLF+ macrophages are more variable across tissues and exhibit more sex differences in gene expression than the other two subsets, which might impact their function. Another recent analysis of human monocytes, macrophages and DCs from healthy tissues as well as inflamed tissues and tumors, revealed broad conservation of subsets across tissues, albeit with some tissue-specific features, including 3 subsets of macrophages in lung, colon, liver, breast, stomach, and pancreatic tumors (26**). The study also suggested that most tumor-associated macrophages are monocyte-derived.
Within an organ, micro-anatomic differences can underlie macrophage diversity. For instance, in the mouse sciatic nerve, two subsets of macrophages with similar developmental origins occupy distinct niches in the epineurium and endoneurium and respond differently to nerve injury (27). Similarly, in a mouse model of nonalcoholic steatohepatitis (NASH), diet-induced changes in liver macrophage subsets include the accumulation of monocyte-derived cells that resemble embryo-derived Kupffer cells (KCs) and occupy the KC niche in hepatic sinusoids, as well as a monocyte-derived subset with a distinct epigenetic landscape that is located around the central and portal veins (28). Another recent study demonstrated that KCs receive key signals from hepatic stellate cells, whereas lipid-associated macrophages are induced by lipid exposure near the bile ducts (29**).
Heterogeneity has also been observed among monocytes. 3 major subsets of hierarchically-related monocytes have previously been characterized in mice and humans – classical or inflammatory monocytes (Ly6Chi in mice, CD14+ CD16− in humans) that enter tissues and give rise to macrophages and monocyte-derived DCs (moDCs), intermediate monocytes (Ly6Cint in mice, CD14+ CD16+ in humans), and non-classical or patrolling monocytes (Ly6Clo in mice, CD14− CD16+ in humans) that remain in the circulation to maintain the vasculature. However, recent studies have demonstrated additional heterogeneity within these subsets. For instance, one CyTOF study of mouse blood monocytes revealed 6 classical, 3 intermediate and 11 non-classical monocyte clusters (30).
Studies of bone marrow monocytes have shed some light on the role of ontogeny in monocyte heterogeneity. In mouse bone marrow, CXCR4 can be used as a marker to distinguish between pre-monocytes (CXCR4hi) and mature monocytes (CXCR4lo) (31). Moreover, 2 independent pathways originating from GMPs and monocyte-DC progenitors (MDPs) yield classical monocytes with distinct neutrophil-like and DC-like gene expression signatures (NeuMo and DCMo, respectively) in steady-state mouse bone marrow (32, 33). Both GMP- and MDP-derived classical monocytes can yield macrophages, but only MDPs give rise to moDCs (Figure 3A), and production of monocytes via the GMP and MDP pathways can be independently controlled by microbial stimuli (33). Monocytes with neutrophil and DC gene signatures have also been observed in human and mouse lung tumors (11).
Figure 3. Monocyte and DC differentiation and diversity.

(A) Monocytes arise from GMPs and MDPs, which also produce neutrophils and cDCs/pDCs, respectively. Classical monocytes produced by GMPs have a neutrophil gene signature (NeuMos), whereas their MDP-derived counterparts (DCMos) express MHCII and can give rise to moDCs. (B) cDCs (cDC1s and CDC2s) arise from CDPs via pre-cDCs, although DC3 cells (a cDC2 subset) are thought to arise independently of CDPs from an early myeloid progenitor. pDCs arise from CDPs via pre-pDCs or from the lymphoid lineage via BLPs.
GMP – granulocyte-monocyte progenitor; MDP – monocyte-DC progenitor; MP – monocyte progenitor (GMP-derived); cMoP – common monocyte progenitor (MDP-derived); CDP – common DC progenitor; CLP – common lymphoid progenitor; BLP – B cell-biased lymphoid progenitor.
Additional heterogeneity emerges when monocytes are activated by microbial components and inflammatory mediators in the circulation or in tissues. For instance, a subset of classical monocytes expressing the IL-7 receptor subunit CD127 has been observed in the bronchoalveolar lavage fluid of COVID-19 patients and in the circulation and synovium of rheumatoid arthritis patients (34). CD127hi monocytes are less inflammatory than their CD127lo counterparts due to IL-7-induced STAT5-dependent chromatin remodeling, which suppresses inflammatory gene expression.
The Heterogeneity and Origins of Dendritic Cells
Dendritic cells (DCs) can be classified as three main subtypes: monocyte-derived DCs (moDCs, discussed above), plasmacytoid DCs (pDCs) and professional antigen-presenting conventional DCs (cDCs). pDCs are notably efficient at producing type I interferon during viral infections, but they can also present antigen to activate T cells. This functional diversity has recently been reported to reflect sequential phases of activation of the same cells in different locations within the mouse spleen (type I IFN production in the marginal zone, then antigen presentation in the T cell zone), rather than functional specialization of different subsets of pDCs (35). cDCs can be subdivided into two major subsets based on cell surface markers, the transcription factors that control their differentiation, and their functions (36). cDC1s (mouse – XCR1; human – XCR1, CLEC9A, CADM1) depend on IRF8 and BATF3 for their differentiation and have the capacity to present and cross-present antigens to CD8+ T cells and thereby induce cytotoxic responses against intracellular pathogens and tumors. cDC2s (mouse – CD11b, CD172a; human – CD1C, CD172a) share some genes with monocytes, differentiate under the control of IRF4, and present antigen to CD4+ T cells, thereby playing key roles in immune defense against extracellular pathogens and allergens.
Moreover, two subsets of cDC2s have been reported in human peripheral blood – DC2 and DC3 – in addition to cDC1s, pDCs, DC precursors, and DCs of monocytic origin (37, 38). Additional cDC2 heterogeneity has been reported in human and mouse lymphoid tissues, which possess cDC2 cells expressing T-bet (cDC2A) that are absent from the blood and express transcripts associated with tissue repair, as well as cDC2 cells expressing RORγt (cDC2B) that are transcriptionally related to blood DC3 and have an increased proinflammatory potential (39). cDC2A and cDC2B appear to acquire their distinct phenotypes in the periphery in response to microenvironmental signals. A CD14+ cDC2 subset related to DC3s that exhibits pro-inflammatory functions has also been reported to accumulate in the blood of systemic lupus erythematosus patients (40).
Subpopulations displaying hybrid phenotypes have also been described, including a subset of mouse splenic DCs hypothesized to represent a transitional state between pre-DCs to cDCs, and human Axl+ Siglec6+ DCs (AS DCs), which have cDC2 and pDC features and are related to mouse transitional DCs (tDCs) that accumulate in the lung during influenza infection (37, 41, 42*). Additionally, inflammatory cDC2s (inf-cDC2s), which migrate efficiently to draining lymph nodes and prime CD4+ and CD8+ T cell responses in several inflammatory disease models, have similarities with monocytes and cDC1s (43), and merocytic DCs (mcDCs), a subset with the ability to reverse T cell anergy when presenting peptides derived from apoptotic bodies, exhibit a unique transcriptomic and metabolic signature with characteristics of both cDC1s and cDC2s (44). In contrast, mature DCs enriched in immunoregulatory molecules (mregDCs) have been observed in human and mouse non-small-cell lung cancers and can arise from both cDC1s and cDC2s upon uptake of tumor antigens (45).
pDCs and cDCs originate in the bone marrow from common DC progenitors (CDPs), which develop from MDPs (Figure 3B). cDCs derive from committed pre-cDC precursors that leave the bone marrow and traffic through the bloodstream to seed lymphoid organs and peripheral tissues, where they finally differentiate into DCs. Pre-cDCs include already committed pre-cDC1s and pre-cDC2s (46). Pre-pDCs have also been identified, but while they had previously been thought to originate from CDPs, recent data also indicates a lymphoid origin via a Ly6D+ B cell-biased lymphoid progenitor (BLP) with differentiation independent of the myeloid lineage (47). Recent studies also provided evidence that DC3s develop along an IRF8low trajectory from a DC3-restricted progenitor that arises from an early myeloid progenitor, in contrast to pDCs, cDC1s and DC2s, which derive from IRF8high progenitors (38, 48).
Interestingly, as discussed above, moDCs have distinct origins from cDCs and pDCs, but a scRNAseq study reported 7 clusters of moDCs differentiated in vitro from human peripheral blood CD14+ monocytes, including 5 that resembled cDC2s (three DC2-like and two DC3-like) (49*). Thus, cells with distinct origins can ultimately have similar characteristics, presumably due to overlapping differentiation programs.
Conclusion
Collectively, a growing body of literature has begun to reveal the mechanisms underlying myeloid heterogeneity, including distinct developmental origins and diverse microenvironmental signals. Additional mechanisms that require further attention include the impact of sex differences – both sex chromosomes and sex hormones, as well as aging – both cell-intrinsic mechanisms such as clonal hematopoiesis, and the extrinsic influence of the more inflammatory microenvironment during “inflammaging”. Moreover, plasticity is a key feature of myeloid cells so phenotypically distinct cells, especially those that arise under disease conditions, may represent different and potentially reversible activation states rather than distinct subsets. It may also be possible to override the impact of origins and memory of prior exposures. Thus, one important priority for the field is to define how subset identity is maintained at the level of chromatin organization and whether deleterious features could be manipulated by selective targeting for therapeutic purposes in disease-specific contexts.
Key points.
Single cell profiling has revealed that neutrophils, monocytes, macrophages and DCs are more heterogeneous than previously appreciated.
Heterogeneity is in part a reflection of diverse origins, including spatial and temporal variation in subset production during development and after birth, and distinct progenitors and differentiation programs.
Heterogeneity can also arise due to variation in microenvironmental signals between and within organs, and during homeostasis versus in response to infection, inflammation or tumors.
Financial Support and Sponsorship
This work was supported by Ramón y Cajal contract (RYC-2017-22895) from the Ministerio de Ciencia, Innovación y Universidades, Spain, a grant (RTI2018-093426-B-100) from the Ministerio de Ciencia, Innovación y Universidades, Spain and the European Fund for Regional Development, and a grant (AICO/2021/350) from the Conselleria d’Innovació, Universitats, Ciència i Societat Digital per a la Promoció de la Investigació Científica, el Desenvolupament Tecnològic i la Innovació a la Comunitat Valenciana, Spain (AY); and a grant from the National Institutes of Health, USA (R01 AI134987) and funds from the Board of Governors Regenerative Medicine Institute at Cedars-Sinai Medical Center (HSG).
Footnotes
Conflicts of interest
The authors declare that they have no conflicts of interest.
References and Recommended Reading
- 1.Ng LG, Ostuni R, Hidalgo A. Heterogeneity of neutrophils. Nat Rev Immunol. 2019;19(4):255–65. [DOI] [PubMed] [Google Scholar]
- 2.Kwok I, Becht E, Xia Y, Ng M, Teh YC, Tan L, et al. Combinatorial Single-Cell Analyses of Granulocyte-Monocyte Progenitor Heterogeneity Reveals an Early Uni-potent Neutrophil Progenitor. Immunity. 2020;53(2):303–18 e5. [DOI] [PubMed] [Google Scholar]
- 3.Muench DE, Olsson A, Ferchen K, Pham G, Serafin RA, Chutipongtanate S, et al. Mouse models of neutropenia reveal progenitor-stage-specific defects. Nature. 2020;582(7810):109–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Evrard M, Kwok IWH, Chong SZ, Teng KWW, Becht E, Chen J, et al. Developmental Analysis of Bone Marrow Neutrophils Reveals Populations Specialized in Expansion, Trafficking, and Effector Functions. Immunity. 2018;48(2):364–79 e8. [DOI] [PubMed] [Google Scholar]
- 5.Dinh HQ, Eggert T, Meyer MA, Zhu YP, Olingy CE, Llewellyn R, et al. Coexpression of CD71 and CD117 Identifies an Early Unipotent Neutrophil Progenitor Population in Human Bone Marrow. Immunity. 2020;53(2):319–34 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Xie X, Shi Q, Wu P, Zhang X, Kambara H, Su J, et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat Immunol. 2020;21(9):1119–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.**.Cugurra A, Mamuladze T, Rustenhoven J, Dykstra T, Beroshvili G, Greenberg ZJ, et al. Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and CNS parenchyma. Science. 2021;373(6553):eabf7844. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that the CNS is supplied with neutrophils and monocytes from adjacent bone marrow niches in the skull in the steady-state and after CNS injury or neuroinflammation, which provides new insights into mechanisms of CNS inflammation.
- 8.Ballesteros I, Rubio-Ponce A, Genua M, Lusito E, Kwok I, Fernandez-Calvo G, et al. Co-option of Neutrophil Fates by Tissue Environments. Cell. 2020;183(5):1282–97 e18. [DOI] [PubMed] [Google Scholar]
- 9.Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell. 2009;16(3):183–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Tsuda Y, Takahashi H, Kobayashi M, Hanafusa T, Herndon DN, Suzuki F. Three different neutrophil subsets exhibited in mice with different susceptibilities to infection by methicillin-resistant Staphylococcus aureus. Immunity. 2004;21(2):215–26. [DOI] [PubMed] [Google Scholar]
- 11.Zilionis R, Engblom C, Pfirschke C, Savova V, Zemmour D, Saatcioglu HD, et al. Single-Cell Transcriptomics of Human and Mouse Lung Cancers Reveals Conserved Myeloid Populations across Individuals and Species. Immunity. 2019;50(5):1317–34 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.**.Crainiciuc G, Palomino-Segura M, Molina-Moreno M, Sicilia J, Aragones DG, Li JLY, et al. Behavioural immune landscapes of inflammation. Nature. 2022;601(7893):415–21. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study of neutrophil shape and mobility sheds new light on the functional diversity and dynamics of neutrophils during inflammation.
- 13.Deniset JF, Surewaard BG, Lee WY, Kubes P. Splenic Ly6G(high) mature and Ly6G(int) immature neutrophils contribute to eradication of S. pneumoniae. J Exp Med. 2017;214(5):1333–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bogoslowski A, Wijeyesinghe S, Lee WY, Chen CS, Alanani S, Jenne C, et al. Neutrophils Recirculate through Lymph Nodes to Survey Tissues for Pathogens. J Immunol. 2020;204(9):2552–61. [DOI] [PubMed] [Google Scholar]
- 15.Lok LSC, Dennison TW, Mahbubani KM, Saeb-Parsy K, Chilvers ER, Clatworthy MR. Phenotypically distinct neutrophils patrol uninfected human and mouse lymph nodes. Proc Natl Acad Sci U S A. 2019;116(38):19083–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Fites JS, Gui M, Kernien JF, Negoro P, Dagher Z, Sykes DB, et al. An unappreciated role for neutrophil-DC hybrids in immunity to invasive fungal infections. PLoS Pathog. 2018;14(5):e1007073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.*.Grieshaber-Bouyer R, Radtke FA, Cunin P, Stifano G, Levescot A, Vijaykumar B, et al. The neutrotime transcriptional signature defines a single continuum of neutrophils across biological compartments. Nat Commun. 2021;12(1):2856. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study proposes a continuum of neutrophil states called “neutrotime” to explain how diverse neutrophil subsets can arise without committed developmental branches.
- 18.Masterson JC, Menard-Katcher C, Larsen LD, Furuta GT, Spencer LA. Heterogeneity of Intestinal Tissue Eosinophils: Potential Considerations for Next-Generation Eosinophil-Targeting Strategies. Cells. 2021;10(2):426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Oetjen LK, Noti M, Kim BS. New insights into basophil heterogeneity. Semin Immunopathol. 2016;38(5):549–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Vivanco Gonzalez N, Oliveria JP, Tebaykin D, Ivison GT, Mukai K, Tsai MM, et al. Mass Cytometry Phenotyping of Human Granulocytes Reveals Novel Basophil Functional Heterogeneity. iScience. 2020;23(11):101724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cildir G, Yip KH, Pant H, Tergaonkar V, Lopez AF, Tumes DJ. Understanding mast cell heterogeneity at single cell resolution. Trends Immunol. 2021;42(6):523–35. [DOI] [PubMed] [Google Scholar]
- 22.Bleriot C, Chakarov S, Ginhoux F. Determinants of Resident Tissue Macrophage Identity and Function. Immunity. 2020;52(6):957–70. [DOI] [PubMed] [Google Scholar]
- 23.Guilliams M, Thierry GR, Bonnardel J, Bajenoff M. Establishment and Maintenance of the Macrophage Niche. Immunity. 2020;52(3):434–51. [DOI] [PubMed] [Google Scholar]
- 24.Bekkering S, Dominguez-Andres J, Joosten LAB, Riksen NP, Netea MG. Trained Immunity: Reprogramming Innate Immunity in Health and Disease. Annu Rev Immunol. 2021;39:667–93. [DOI] [PubMed] [Google Scholar]
- 25.**.Dick SA, Wong A, Hamidzada H, Nejat S, Nechanitzky R, Vohra S, et al. Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles. Sci Immunol. 2022;7(67):eabf7777. [DOI] [PubMed] [Google Scholar]; This study demonstrates the existence of 3 conserved macrophage subtypes with distinct features and origins across diverse organs, including the heart, liver, lung, kidney and brain.
- 26.**.Mulder K, Patel AA, Kong WT, Piot C, Halitzki E, Dunsmore G, et al. Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease. Immunity. 2021;54(8):1883900 e5. [DOI] [PubMed] [Google Scholar]; This study profiled human monocytes, macrophages and DCs from 41 datasets, including 13 healthy tissues plus inflamed tissues and tumors, revealing broad conservation of subsets across tissues, including 3 subsets of macrophages in lung, colon, liver, breast, stomach, and pancreatic tumors.
- 27.Ydens E, Amann L, Asselbergh B, Scott CL, Martens L, Sichien D, et al. Profiling peripheral nerve macrophages reveals two macrophage subsets with distinct localization, transcriptome and response to injury. Nat Neurosci. 2020;23(5):676–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Seidman JS, Troutman TD, Sakai M, Gola A, Spann NJ, Bennett H, et al. Niche-Specific Reprogramming of Epigenetic Landscapes Drives Myeloid Cell Diversity in Nonalcoholic Steatohepatitis. Immunity. 2020;52(6):1057–74 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.**.Guilliams M, Bonnardel J, Haest B, Vanderborght B, Wagner C, Remmerie A, et al. Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches. Cell. 2022;185(2):379–96 e38. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study reveals how diverse populations of liver macrophages acquire and maintain their distinct phenotypes under the influence of microenvironmental cues. Kupffer cells rely on signals from hepatic stellate cells, whereas lipid-associated macrophages located near the bile ducts are induced by lipid exposure.
- 30.Roberts ME, Barvalia M, Silva J, Cederberg RA, Chu W, Wong A, et al. Deep Phenotyping by Mass Cytometry and Single-Cell RNA-Sequencing Reveals LYN-Regulated Signaling Profiles Underlying Monocyte Subset Heterogeneity and Lifespan. Circ Res. 2020;126(10):e61–e79. [DOI] [PubMed] [Google Scholar]
- 31.Chong SZ, Evrard M, Devi S, Chen J, Lim JY, See P, et al. CXCR4 identifies transitional bone marrow premonocytes that replenish the mature monocyte pool for peripheral responses. J Exp Med. 2016;213(11):2293–314. [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. 2020;367(6479):eaaw3381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yanez A, Coetzee SG, Olsson A, Muench DE, Berman BP, Hazelett DJ, et al. GranulocyteMonocyte Progenitors and Monocyte-Dendritic Cell Progenitors Independently Produce Functionally Distinct Monocytes. Immunity. 2017;47(5):890–902 e4, e20211191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhang B, Zhang Y, Xiong L, Li Y, Zhang Y, Zhao J, et al. CD127 imprints functional heterogeneity to diversify monocyte responses in inflammatory diseases. J Exp Med. 2022;219(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Abbas A, Vu Manh TP, Valente M, Collinet N, Attaf N, Dong C, et al. The activation trajectory of plasmacytoid dendritic cells in vivo during a viral infection. Nat Immunol. 2020;21(9):983–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Anderson DA 3rd, Dutertre CA, Ginhoux F, Murphy KM. Genetic models of human and mouse dendritic cell development and function. Nat Rev Immunol. 2021;21(2):101–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Villani AC, Satija R, Reynolds G, Sarkizova S, Shekhar K, Fletcher J, et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science. 2017;356(6335):eaah4573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cytlak U, Resteu A, Pagan S, Green K, Milne P, Maisuria S, et al. Differential IRF8 Transcription Factor Requirement Defines Two Pathways of Dendritic Cell Development in Humans. Immunity. 2020;53(2):353–70 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Brown CC, Gudjonson H, Pritykin Y, Deep D, Lavallee VP, Mendoza A, et al. Transcriptional Basis of Mouse and Human Dendritic Cell Heterogeneity. Cell. 2019;179(4):846–63 e24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dutertre CA, Becht E, Irac SE, Khalilnezhad A, Narang V, Khalilnezhad S, et al. Single-Cell Analysis of Human Mononuclear Phagocytes Reveals Subset-Defining Markers and Identifies Circulating Inflammatory Dendritic Cells. Immunity. 2019;51(3):573–89 e8. [DOI] [PubMed] [Google Scholar]
- 41.Leylek R, Alcantara-Hernandez M, Lanzar Z, Ludtke A, Perez OA, Reizis B, et al. Integrated Cross-Species Analysis Identifies a Conserved Transitional Dendritic Cell Population. Cell Rep. 2019;29(11):3736–50 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.*.Lukowski SW, Rodahl I, Kelly S, Yu M, Gotley J, Zhou C, et al. Absence of Batf3 reveals a new dimension of cell state heterogeneity within conventional dendritic cells. iScience. 2021;24(5):102402. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study describes a subset of mature cDC1 cells expressing surface markers characteristic of cDC2 cells.
- 43.Bosteels C, Neyt K, Vanheerswynghels M, van Helden MJ, Sichien D, Debeuf N, et al. Inflammatory Type 2 cDCs Acquire Features of cDC1s and Macrophages to Orchestrate Immunity to Respiratory Virus Infection. Immunity. 2020;52(6):1039–56 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Audiger C, Fois A, Thomas AL, Janssen E, Pelletier M, Lesage S. Merocytic Dendritic Cells Compose a Conventional Dendritic Cell Subset with Low Metabolic Activity. J Immunol. 2020;205(1):121–32. [DOI] [PubMed] [Google Scholar]
- 45.Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel J, et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature. 2020;580(7802):257–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Cabeza-Cabrerizo M, van Blijswijk J, Wienert S, Heim D, Jenkins RP, Chakravarty P, et al. Tissue clonality of dendritic cell subsets and emergency DCpoiesis revealed by multicolor fate mapping of DC progenitors. Sci Immunol. 2019;4(33):eaaw1941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dress RJ, Dutertre CA, Giladi A, Schlitzer A, Low I, Shadan NB, et al. Plasmacytoid dendritic cells develop from Ly6D(+) lymphoid progenitors distinct from the myeloid lineage. Nat Immunol. 2019;20(7):852–64. [DOI] [PubMed] [Google Scholar]
- 48.Bourdely P, Anselmi G, Vaivode K, Ramos RN, Missolo-Koussou Y, Hidalgo S, et al. Transcriptional and Functional Analysis of CD1c(+) Human Dendritic Cells Identifies a CD163(+) Subset Priming CD8(+)CD103(+) T Cells. Immunity. 2020;53(2):335–52 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.*.Gao Y, Li H, Li Z, Xie L, Liu X, Huang Z, et al. Single-Cell Analysis Reveals the Heterogeneity of Monocyte-Derived and Peripheral Type-2 Conventional Dendritic Cells. J Immunol. 2021;207(3):837–48. [DOI] [PubMed] [Google Scholar]; This study profiles the transcriptomes of DCs derived from human peripheral blood monocytes using GM-CSF, which are the most readily available human DC source. The authors report 7 subtypes of moDCs, including 5 with features of cDC2s, which has implications for the use of DCs for cell therapies.
