Monocyte, Macrophage, and Dendritic Cell

1 downloads 0 Views 8MB Size Report
the vertex of the two-dimensional plot. These have ...... Yona S, Kim KW, Wolf Y, Mildner A, Varol D, Breker M, Strauss- ... Bain CC, Mowat AM. .... CD14, CD16 and HLA-DR reliably identifies human monocytes ..... SE, Rogers NC, Moncaut N, Carvajal JJ, Reis e Sousa C. 2013. .... Stanley ER, Randolph GJ, Merad M. 2006.
Monocyte, Macrophage, and Dendritic Cell Development: the Human Perspective MATTHEW COLLIN1 and VENETIA BIGLEY1 1

Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom

ABSTRACT The maintenance of monocytes, macrophages, and dendritic cells (DCs) involves manifold pathways of ontogeny and homeostasis that have been the subject of intense study in recent years. The concept of a peripheral mononuclear phagocyte system continually renewed by blood-borne monocytes has been modified to include specialized DC pathways of development that do not involve monocytes, and longevity through self-renewal of tissue macrophages. The study of development remains difficult owing to the plasticity of phenotypes and misconceptions about the fundamental structure of hematopoiesis. However, greater clarity has been achieved in distinguishing inflammatory monocyte-derived DCs from DCs arising in the steady state, and new concepts of conjoined lymphomyeloid hematopoiesis more easily accommodate the shared lymphoid and myeloid phenotypes of some DCs. Cross-species comparisons have also yielded coherent systems of nomenclature for all mammalian monocytes, macrophages, and DCs. Finally, the clear relationships between ontogeny and functional specialization offer information about the regulation of immune responses and provide new tools for the therapeutic manipulation of myeloid mononuclear cells in medicine.

INTRODUCTION Monocytes, macrophages, and dendritic cells (DCs) are populations of myeloid mononuclear cells (MMCs) that provide critical sensing functions in innate immunity and a bridge to adaptive immunity through antigen presentation. They also perform important effector functions and contribute to chronic inflammation and healing. Collectively they have been described as the “mononuclear phagocyte system” (MPS) (1). As originally conceived, the MPS had a single blood-borne precursor, the monocyte. It is now appreciated that the development

and homeostasis of MMCs is considerably more complex. In this chapter, we discuss the ontogeny of these diverse cells and reflect on ways in which ontogeny is linked to functional specialization, plasticity, and immune regulation.

MYTHS, CONTROVERSIES, AND OTHER DIFFICULTIES Resolving the developmental relationships between monocytes, macrophages, and DCs and their respective roles in immunity are challenges that continue to attract strong opinion. For many years, the MPS model was widely accepted and both resident histiocytes and myeloid inflammatory infiltrates were presumed to be populations derived from monocytes (Fig. 1A). The ability of human monocytes to take on a wide range of phenotypes in vitro appeared to support this notion (2, 3).

DCs: a Hematopoietic Lineage The discovery of DCs posed a problem for the classic MPS model (4): if they were included as members of the MPS, this implied monocyte origin, promulgating the Received: 16 July 2015, Accepted: 19 April 2016, Published: 16 September 2016 Editor: Siamon Gordon, Oxford University, Oxford, United Kingdom Citation: Collin M, Bigley V. 2016. Monocyte, macrophage, and dendritic cell development: the human perspective. Microbiol Spectrum 4(5):MCHD-0015-2015. doi:10.1128/microbiolspec .MCHD-0015-2015. Correspondence: Matthew Collin, [email protected] © 2016 American Society for Microbiology. All rights reserved.

ASMscience.org/MicrobiolSpectrum

1 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Collin and Bigley

FIGURE 1 Steady-state MMC homeostasis. (A) Classical mononuclear phagocyte model showing the development of LCs, DCs, and macrophages from a common monocyte precursor. (B) Current model of DC, monocyte, and macrophage development in which DCs develop through a discrete hematopoietic lineage arising from a bone marrowresident restricted DC precursor, the CDP. CDPs give rise to pDCs and a myeloid pre-DC population that differentiates into two myeloid DCs (cDC1 and cDC2). It is not known if the pre-DC or cDC1 and cDC2 circulating in the blood give rise to tissue populations of cDC1 and cDC2. Monocytes also contribute to steady-state populations of monocyte-derived macrophages and DCs especially in the skin, gut, and lung. These can be clearly distinguished from the CDP-derived DCs and resident macrophages. Resident macrophages are originally derived from prenatal hematopoiesis. It is unknown whether monocytederived cells can contribute to long-term resident macrophages. cMoP, common monocyte progenitor; MAC, macrophage; mo, monocyte.

2

ASMscience.org/MicrobiolSpectrum Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Monocyte, Macrophage, and Dendritic Cell Development

view that DCs were just a type of specialized macrophage (5). If one accepted their uniqueness, then this suggested the existence of a distinct DC lineage that was not consistent with a unified MPS. The heritage of DCs took some years to emerge following their discovery, but it is now accepted by most that DCs emerge from the bone marrow as a defined hematopoietic lineage that is distinct from monocytes (6) (Fig. 1B). While monocyte function overlaps the remit of DCs in inflammation, it is clear that in the steady state easily distinguishable populations of DCs and monocytes, or their derivatives, exist in the blood and tissues of humans and other mammals (7–11). The significance of a distinct DC lineage in the induction of immunity is still incompletely understood, as although DCs are potent at stimulating naive T cells, they are relatively rare, especially during inflammation, when monocytederived, highly proficient antigen-presenting cells are rapidly formed (12). While monocyte function may be sufficient to resist infection at some level, DCs presumably improve the fitness of the immune system to respond rapidly, to generate anamnestic responses, or to combat a small number of highly virulent pathogens exerting strong selective pressure. The nonredundant role of DCs in infection and evidence of their tolerogenic functions are established in a number of mouse models (13, 14).

Primitive Macrophages A second, major challenge to the MPS model was the more recent discovery that the majority of histiocytes, or tissue-resident macrophages, are extremely stable populations that do not require replenishment by monocytes or any other precursor (15, 16). In the absence of inflammatory challenge, microglia, Langerhans cells (LCs), Kupffer cells, and alveolar, peritoneal, and splenic macrophages persist from fetal or even embryonic hematopoiesis (12, 17, 18). Notable exceptions are the contributions of monocyte-derived cells to skin, gut, and interstitial lung macrophages (10, 19, 20). Significant import has been attached to the primitive origin of tissue macrophages, and recent studies in the heart suggest that they have a much more prominent role in repair than recently recruited monocyte-derived cells (21). It is not clear whether monocyte-independent routes of macrophage differentiation can also persist in some form after birth. Given that hematopoietic stem cells (HSCs) are detectable in the peripheral tissues of mice (22), it is hard to exclude the occurrence of a direct route of macrophage differentiation from an early hematopoietic precursor, residing in adult tissues.

PITFALLS IN THE STUDY OF MMCs This narrative illustrates a number of generic challenges in the study of monocytes, macrophages, and DCs. First, it is important to distinguish between the steady state and inflammation. There is no a priori reason to suppose that inflammatory precursors and their progeny are related to steady-state populations of MMCs. Indeed, temporal and special separation of quiescent and inflammatory pathways of differentiation may provide critical aspects of immune regulation. Major questions remain unsolved, particularly in humans, concerning the functional differences between resident and recruited cells and their respective fates after inflammation has resolved. Second, overreliance on a small number of surface antigens to identify a particular cell type can be misleading. The use of CD11c and major histocompatibility complex class II (MHC-II) to define murine DCs has been criticized (5). In humans, it was already well known that CD11c and MHC-II are highly expressed by both monocytes and DCs and cannot be used to separate them in the blood, although CD11c defines different populations of dermal DCs and macrophages quite clearly in tissues (8). This issue has been partly circumvented by the discovery of more-restricted antigens such as the suite of “blood DC antigens,” or BDCA-1 to -4, in humans (23, 24) and, more recently, new DC and macrophage signposts such as CLEC9A (C-type lectin domain family 9 member A), XCR1 (chemokine XC receptor 1), SIRPα (signal regulatory protein α), and MerTK (MER tyrosine kinase) identified by unbiased computational approaches to classification (25, 26). Third, the plasticity of MMCs to respond to environmental cues means that different anatomical sites are likely to induce specific phenotypes (27). Furthermore, this plasticity inflates the problems already alluded to that inflammation and the steady state are not equivalent and that individual surface antigens may be fickle. A cogent argument has been made to mitigate these problems by using ontogeny as the basis of MMC classification (28). This approach has many attractions, and although definitive ontological experiments are difficult to perform in humans, the concepts developed in mice can often translate to humans. As recent data show, the prenatal development of MMCs is an important dimension to consider, in addition to the lineages that arise later from definitive HSCs (12, 17).

THE REPERTOIRE OF HUMAN MONOCYTES, MACROPHAGES, AND DCs Monocytes Monocytes are the archetypal MMCs, constituting ∼10% of human peripheral blood mononuclear cells.

ASMscience.org/MicrobiolSpectrum

3 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Collin and Bigley

They are blood borne by convention, although recent studies in humans and mice describe steady-state populations of tissue monocytes or monocyte-derived cells (9, 19, 20). Monocytes have a plethora of functions in inflammation and rapidly differentiate into cells with DC-like and macrophage phenotypes that can be difficult to separate from resident populations. Recent transcriptomic profiling experiments highlight the complexity of monocyte differentiation potential (29). Human monocytes are heterogeneous in their expression of CD14 and CD16 (Fc receptor III), and this has been used to divide them into subpopulations (30). CD14

and CD16 are both continuously distributed (Fig. 2), but the majority of monocytes are CD14+ and are defined as classical or inflammatory monocytes. Most of the precursor functions defined in vitro, including the formation of monocyte-derived DCs and macrophages, arise from classical monocytes (29). Some studies ascribe distinct functions to “intermediate” CD14+CD16+ monocytes at the vertex of the two-dimensional plot. These have the highest MHC-II expression and cytokine production and may be derived by activation of classical monocytes. Nonclassical CD16+ monocytes also express high MHC-II and costimulatory antigens, leading some

FIGURE 2 Human blood monocytes and DCs. Gating strategy to identify human monocytes and DCs in peripheral blood. Monocytes and DCs are all found in the HLA-DR+, lineage-negative compartment. CD14 versus CD16 displays monocyte subsets and double-negative DC populations. These may be separated in a variety of ways. Here the markers CD123 and CD11c are used to define pDCs and myeloid DCs. The latter can be separated into cDC1 and cDC2 using CD141 and CD1c, respectively.

4

ASMscience.org/MicrobiolSpectrum Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Monocyte, Macrophage, and Dendritic Cell Development

authors to regard them as DCs (24). However, transcriptional profiling and unsupervised hierarchical clustering is unequivocal that all monocytes cluster independently of DCs (9, 31). Within the CD16+ subset a population expressing the antigen 6-sulfo LacNAc (SLAN-DCs) is reported to secrete large amounts of tumor necrosis factor α, interleukin-1β (IL-1β), and IL-12 and to respond rapidly to inflammatory stimuli (32). Other studies suggest that CD16+ monocytes, including SLAN+ cells, have low inflammatory activity and are homologous to murine Gr-1/Ly6Clo “patrolling” monocytes (33). Inconsistencies may have arisen through different gating of the CD14 versus CD16 plot and inclusion or exclusion of the CD14+CD16+ intermediate cells, which have a highly activated phenotype. Anatomically, it appears that nonclassical monocytes are well positioned to infiltrate tissues or to control diapedesis across the endothelium; the importance of both of these potential functions remains to be tested in humans (34). The origin of human nonclassical monocytes remains controversial. Kinetic studies in the mouse suggest that they are derived from classical monocytes (35), although this is recently disputed (36). Human CD16+ monocytes also have higher expression of the nuclear factor Nur77, proposed to control murine nonclassical monocyte differentiation (37). There is a prominent literature in humans describing abnormalities of monocyte development in disease, notably the appearance of “sick” monocytes with lower MHC-II expression and impaired antimicrobial functions (38). Variation in the CD14/CD16 ratio due to expansion or contraction of either subset is also associated with many different conditions. Of note, CD16+ monocytes are highly sensitive to depletion following treatment with corticosteroids (39). The appearance of monocytic “myeloid-derived suppressor cells” that can inhibit T-cell responses is associated with cancer and chronic infection (40). These demographic changes may simply represent a “left shift” in myelopoiesis, but nonetheless this appears to provide immune regulation under conditions of stress or chronic inflammation. Finally, heterogeneous expression of CD68, CD163, and CD202b (Tie2; angiopoietin receptor) has been noted among human monocytes. Tie2+ monocytes are found throughout classical, intermediate, and nonclassical populations and are implicated in tumor angiogenesis (41).

Macrophages: Histiocytes Old and New Macrophages are classically tissue-resident cells, or “histiocytes.” Many different specialized populations

of macrophages exist in different sites. Observations from human HSC transplantation and monocytopenic states suggest that as in mice, these are likely to be developmentally old cells that can persist for long periods without replenishment from the bone marrow (7, 42– 44). The label “histiocyte” is perhaps still useful to denote these developmentally older populations from more recently recruited monocyte-derived cells. Location matters, and the tissue environment induces specific gene regulation in resident histiocytes, regardless of their origin (27). Recent advances have led to much clearer definitions between long-term resident histiocytes, DCs, and more transient populations of monocyte-derived cells (Fig. 3). Macrophages are operationally distinguished from DCs by their much lower ability to simulate naive T cells (4). There are also many differences in gene expression (9, 25, 26). Useful markers in human skin such as CD163, factor XIIIA, and LYVE-1 (lymphatic vessel endothelial hyaluronan receptor 1) identify macrophage populations that are completely nonoverlapping with DCs, identified by CD11c or CD1c expression (8, 45). In addition to histiocytes and DCs, populations of monocytederived cells such as CD14+ or “interstitial-type” DCs are also found in human skin. These were originally classified as DCs owing to their ability to migrate from explanted skin (46), but transcriptomic profiling shows close relationships with the human dermal macrophage and mouse monocyte-derived macrophages (9, 10). As predicted, these monocyte-derived macrophages are indeed poor allostimulators, produce IL-10, and can provide regulatory function (47). Confusingly, CD141 may also be induced on monocyte-derived cells, but they remain distinct from CD141+ DCs particularly through the expression of CD14, SIRPα, and other markers (11, 47). In vivo human data show that monocyte-derived macrophages are rapidly repopulated after HSC transplantation, in contrast to resident histiocytes (7, 9). Monocyte-derived cells are also rapidly turned over in murine tissues and may be found in afferent lymph (10, 19, 20). Together, these observations support a model that recently derived monocyte-macrophages are mobile, in contrast to the long-term histiocyte population. Immobility and the progressive accumulation of lipofuscin and pigment that gives macrophages autofluorescence in the flow cytometer are physical signs of longevity that are experimentally tractable to identify histiocytes and to separate them from monocyte-derived cells (7, 9). One difference between mouse and human is that it has been difficult to demonstrate homeostatic proliferation in human macrophage populations. Observations

ASMscience.org/MicrobiolSpectrum

5 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Collin and Bigley

FIGURE 3 Summary of human DC, monocyte, and macrophage origins in relation to steady-state surface markers. The most distinctive DC phenotypes are displayed by pDCs and CD141+ cDC1 cells. There is more overlap between cDC2 and monocyte-derived cells that may coexist in tissues. Monocyte-derived macrophages also share many markers with resident histiocytes. The relative size of arrows from the precursor populations indicates the estimated steady-state flux. CMoP, common monocyte progenitor; mac, macrophage; mo, monocyte.

in quiescent skin suggest that if proliferation does occur, it is less than one-tenth of the level observed in LCs (∼2 to 3%) (7, 43). These observations need to be extended to inflammatory situations such as Th2 environments that have been observed to stimulate murine macrophage proliferation (48). The relatedness of monocytes, monocyte-derived macrophages, and tissue histiocytes by unsupervised clustering of their gene expression signatures is reminiscent of the classic picture of the MPS as a monocytebased system but also strong evidence of the separateness of the DC lineage, members of which cluster very distinctly (9, 31). However, care must be made in extrapolating these static similarities to population dynamics. The plasticity of monocytes allows them take

on genuine DC properties during inflammation (49–51). We concur with others that in experimental work the term “monocyte-derived DC” should be used to describe such populations (12, 28, 52).

Dendritic Cells DCs were first isolated from the mouse spleen as nonadherent branching cells that were 100-fold more potent at activating naive T cells compared with adherent macrophages (4). “Resident” populations of DCs that are derived from blood-borne precursors are found in lymphoid tissues, while “migratory” DCs found in peripheral tissues take up exogenous antigens and migrate to draining lymph nodes, performing a surveillance role (6).

6

ASMscience.org/MicrobiolSpectrum Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Monocyte, Macrophage, and Dendritic Cell Development

For obvious logistical reasons the analysis of human lymphoid and tissue DCs has proceeded more slowly than the study of human blood. In human blood, three populations of DCs are readily described: plasmacytoid DCs (pDCs) and two populations of “myeloid” DCs (Fig. 3). The term “myeloid” is equivalent to “classical” or “conventional” in mice. Its usage in humans is quite specific and based on the expression of antigens typically seen on granulocytes or monocytes, including CD13, CD33, CD11b, and CD11c. These markers are lower or absent on pDCs, which are also morphologically and functionally distinct, providing a major source of type I interferon in response to viral infection (reviewed in reference 53). All DCs carry antigens typically associated with lymphoid differentiation, such as CD4 (54). MHC-II and CD11c are not useful for separating DCs from monocytes in humans, but the series of BDCA antigens is extremely powerful (23, 24). Myeloid DCs separate into a major CD1c+ (BDCA-1) population and a minor CD141+ (BDCA-3) population. The minor population is homologous with the mouse CD8+/CD103+ DC lineage, has high expression of XCR1 and CLEC9A, and has natural cross-presenting capacity (55, 56). In a recent nomenclature system this “conventional” DC lineage has been called cDC1, while human CD1c+ DCs together with mouse conventional CD4+ or CD11b+ DCs are cDC2 (28). An alternative system is simply to name these cells CD141+ myeloid DCs and CD1c+ myeloid DCs, although, as previously noted, CD141+ is quite a promiscuous antigen (30). It has been suggested that more-universal markers are XCR1 and CLEC9A for cDC1 and SIRPα for cDC2 (57). The conservation of pDCs is apparent in many different mammalian species, and in humans they express CD123, CD303 (BDCA-2; CLEC4C), and CD304 (BDCA-4; neuropilin). In tissues it is possible to detect equivalent populations of migratory myeloid DCs. Lymphoid organs also contain CD1c+ and CD141+ DCs in addition to pDCs and LCs in skin-draining locations (58–61).

Langerin-Positive DCs The expression of Langerin on non-LCs was first highlighted in the mouse by several groups working with models of inducible LC depletion, in which Langerin + CD103+EpCAM– DCs were observed in the complete absence of LCs (62). Langerin expression had already been observed in CD8+ lymph node-resident DCs (63), and it became clear that Langerin is expressed by murine CD8+/CD103+ cDC1s. In contrast, a small component of CD1c+ cDC2s express Langerin in human tissues in vivo (64). This population is distinct from LCs, and

the human CD141+ cDC1 population in the steady state, but may be a precursor of LCs after inflammation (65).

Langerhans Cells LCs are an unusual population of migratory myeloid DCs situated in the epidermis and mucosae (66). They express Langerin and CD1a at high levels and contain Birbeck granules by electron microscopy (63). LCs can be stimulated to leave the epidermis, forming migratory DCs detectable in skin-draining lymph nodes. LCs appear to play multiple roles in immunity, including the presentation of lipid antigens (67), inactivation of pathogens (68), and induction of Th17 responses (69) and graft-versus-host disease (70), in addition to the maintenance of tolerance in the steady state (14). LCs are derived from myeloid precursors during fetal life (71, 72) that differentiate under the influence of the colony-stimulating factor-1 receptor (CSF-1R; CD115), cytokine IL-34 (73, 74), and autocrine production of transforming growth factor β (75). Proliferation of epidermal LCs was first reported in humans (76), but elegant experiments in mice subsequently proved that selfrenewal was sufficient to provide local homeostasis (77). LCs were the first example of an MMC that was not continuously replenished from the bone marrow. Limb transplantation and bone marrow failure syndromes confirm their independence of bone marrow-derived precursors in humans (43, 44, 78). Interestingly, it has been difficult to reproduce mouse data showing that LCs survive HSC transplantation, and most are replaced by donor cells within a few months, even with nonmyeloablative conditioning and in the absence of graft-versushost disease (79, 80).

EXPERIMENTAL APPROACHES TO ANALYZING MYELOID DIFFERENTIATION Hematopoiesis has provided one of the seminal model systems of developmental biology. The key concepts are that stem cells are self-renewing and pluripotent and that differentiation proceeds by a progressive restriction of cell potential, through multiple rounds of proliferation. The focus of almost all research has been to define the nodes in this process, the points at which cell potential bifurcates, through the isolation of clonogenic progenitors that give rise to daughter cells each with nonoverlapping developmental potential. Combined with flow cytometry, the major fruit of this labor is the purification of cells with progressively restricted potential, manifest by a number of in vitro or in vivo assays. There are a number of limitations of this approach that are pertinent

ASMscience.org/MicrobiolSpectrum

7 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Collin and Bigley

to unraveling the development and differentiation of MMCs.

Not All Swans Are White Experiments subjecting purified progenitors to any interrogation of their potential can only rule in a particular cell fate potential; they cannot exclude it. Even when a comparator population is used to verify the performance of a readout, a cell that fails in one assay may have the same potential under more sophisticated conditions, for example, the use of stromal cells or new growth factors. This problem has led to the appearance of a number of black swans over the years as new potentials are revealed through improvements in the sensitivity of readouts. Xenografting in immunodeficient mice is no exception, as the expression of cell potential is still limited by an arbitrary range of cross-reactive hematopoietins (81).

Uncertainty Principles In Vitro A further problem in analyzing single cells with dual potential is that it may be difficult or impossible to find a set of conditions that allow simultaneous differentiation along more than one lineage. A progenitor may exist in one of several states of potential, but often this can only be interrogated by collapsing the potential to a singular outcome through defined culture conditions. Critically, attempts to define a “universal” readout are likely to increase the chance of missing a potential outcome and creating an unseen black swan. An interesting metaproblem of the same nature is that antibodies to growth factor receptors are frequently used to sort populations and can potentially interfere with growth factor signaling during the analysis of progeny. Thus, it has been suggested that high expression of CD115 used to isolate restricted DC progenitors may blunt the ability of these cells to develop into monocytes (5). Recent efforts to define DC progenitors in humans have all capitalized on growth factor expression to isolate prospective cell populations (82, 83).

Monocyte and DC Potential The black swan and simultaneous differentiation problems both impede the study of monocyte and DC development in humans. First, conditions permissive for the development of human DCs in vitro and in animal models have only recently been defined (56, 82, 84); and second, the phenotypic readouts are subtle, and robust definitions of what constitutes a DC or monocytederived cell have been hard to formulate, particularly due to the extreme plasticity of monocytes in adopting DC-like phenotypes (51).

In terms of defining the “true” DC potential of an assay, the appearance of pDCs and XCR1+CLEC9A+ myeloid DCs (cDC1) is instrumental. This is because CD1c+ myeloid DCs (cDC2) are close in phenotype to monocyte-derived DCs and it may be difficult to discern if cDC2 phenotype output is simply a reflection of monocyte potential; this is especially true when granulocyte-macrophage CSF (GM-CSF) and IL-4 are present, conditions that promote the formation of monocytederived DCs (85). It is necessary to perform quite detailed transcriptional profiling in order to ascertain the true lineage of culture outputs (82). The ability of CD1c+ DCs to form LC-like cells in response to transforming growth factor β may be useful in distinguishing them from monocyte-derived DCs (65, 86).

THE MYELOID-BASED MODEL OF MOUSE MMC DIFFERENTIATION Many critical details of murine MMC development have recently been elaborated. As alluded to previously, it is now appreciated that primitive myelopoiesis contributes almost exclusively to microglia, which arise directly from yolk sac macrophages, and that LCs and most tissue macrophages also have a primitive origin via fetal liver monocytes (12, 17, 18). Definitive hematopoiesis continues to supply monocytes and DCs throughout the life of an animal, and unraveling a discrete pathway of DC development in the mouse has been a significant advance in the field of hematopoiesis (6). The model that has emerged is firmly myeloid based, in which granulocyte-macrophage progenitors (GMPs) give rise to macrophage and DC progenitors (MDPs), which have lost granulocyte potential (87, 88). MDPs in turn give rise to a monocyte precursor (35) and common DC precursor (CDP) (89, 90). The latter has restricted DC potential and is the key staging post of the DC lineage where monocyte potential should disappear. pDCs are said to arise directly from CDPs, but classical or conventional DCs descend from an intermediate pre-cDC population that can be found in the blood (91, 92) (Fig. 1B). This model appeals to the reductionist by proposing sequential bifurcations of cell potential consistent with textbook models of hematopoiesis. However, a number of inconsistencies are emerging. The MDP has recently come under fresh scrutiny with evidence that its ability to generate pDCs and XCR1+ cDC1s (markers of “true” DC potential) is much less than its monocyte potential and that it may still retain some ability to form granulocytes (93). The identity of the CDP as a clonogenic

8

ASMscience.org/MicrobiolSpectrum Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Monocyte, Macrophage, and Dendritic Cell Development

progenitor has also been questioned, with reports that populations originally classified as CDPs are primed to produce either classical DCs (cDC1 and cDC2) or pDCs according to their level of CLEC9A and CD115 expression (94, 95). Pre-DC function has also been observed in a pDC-like cell that is distinct from the pre-DC (96). Furthermore, the pre-DC itself shows evidence of precommitment to either cDC1 or cDC2 progeny (97). The concept of precommitment or lineage priming is not new to stem cell biology but is at variance with models based on discrete cell fate “decisions” at the nodes of clonogenic progenitors (98). Lentiviral barcoding reveals that lineage priming may bias the entire progeny of early progenitors, with a significant tranche apparently devoted to the generation of DCs (99). A final aspect of the myeloid-based model that has been difficult to assimilate with other mouse and human data is the lack of consideration for lymphoid pathways of DC development (100, 101). The original concept that pDCs might have a lymphoid heritage stems not only from their morphology and surface markers but also from the fact that rearranged IgH and T-cell receptor loci are found at high frequency (102, 103). Rather than “convergent development” from two different branches of hematopoiesis, it appears more likely that lymphoid and myeloid potential are conjoined until late stages of differentiation.

MYELOID AND LYMPHOID OR LYMPHOMYELOID? The terms “myeloid” and “lymphoid” dominate hematopoiesis. “Myeloid” is used here in the broad sense of bone marrow derived, encompassing erythropoiesis, megakaryopoiesis, granulopoiesis, and the production of monocytes and macrophages; indeed anything that does not include small lymphoid cells. In the classical model of hematopoiesis, the earliest dichotomy occurs between myeloid and lymphoid, based on the description of common myeloid (erythro-mega-granulocytemacrophage) and common lymphoid (B and T) progenitors (104, 105). However, in assays that supported both myeloid and lymphoid differentiation, lymphomyeloid progenitors lacking erythro-megakaryocyte potential were observed, suggesting that the specification of lymphoid potential is a more continuous process (81, 106). Many groups have since described “lymphoid-primed multipotent progenitors” (LMPPs) in mice and humans, in support of this concept (85, 107) (Fig. 4). From an epigenetic standpoint, the gradual, rather than abrupt, loss of lymphoid

potential concords with progressive methylation of myeloid regulatory loci (108). From the point of trying to decipher monocyte and DC development, these findings are absolutely key as it is no longer necessary to invoke an unorthodox blending of myeloid and lymphoid potential if instead they coexist. The DC embodies innate myeloid phenotype with an ability to interact with adaptive immune system lymphocytes, and difficulties in mapping its precise origin may be intimately linked to problems with the myeloid-lymphoid dichotomy of classical hematopoiesis. A graphical illustration of this is the structure of hematopoiesis derived from an unbiased analysis of transcription factor expression by the “known” hematopoietic lineages and their intermediates: this places the DC lineage as neither myeloid nor lymphoid but arising directly from HSCs (109).

HUMAN MONOCYTE AND DC DIFFERENTIATION Serious analysis of human hematopoiesis began with in vitro colony-forming assays defining erythroid-, megakaryocytic-, granulocyte-, and “macrophage”forming units, thus providing the first example of readouts that could interrogate either granulocyte or monocyte potential (110). CD34 is a key marker of the human progenitor/stem cell compartment. Most multilineage potential is contained within the CD38lo fraction of CD34+ cells, while CD38+ cells contain committed cells. The CD38+CD10+ fraction contains lymphoid potential, while GMPs fall in the CD38+CD45RA+ fraction. The importance of CSF-1R in identifying progenitors with monocyte but not granulocyte potential was recognized early (111). This population potentially represents MDP-like cells within the GMP, but to our knowledge a human equivalent of the common monocyte progenitor has not been identified. In addition, within the GMP, a discrete population of CD123hi (IL-3 receptor) cells was noted to give rise to pDCs (112). CD123hi cells form a distinct peninsula of the GMP cloud that has been observed in a number of studies (113). Upon in vitro exposure to Flt3 ligand, stem cell factor, GM-CSF, and stromal cells, the CD123hi population of GMPs contains pDC and myeloid cDC1 and cDC2 potential but lacks monocyte potential (82). In this study, molecular profiling was required to differentiate between monocytes and cDC2 because CD1c+ cDC2s can acquire CD14 expression in vitro and CD14 monocytes can differentiate into CD1c+ cDC2-like cells. These results show that good DC potential resides in the

ASMscience.org/MicrobiolSpectrum

9 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Collin and Bigley

FIGURE 4 Revised structure of human hematopoiesis showing common lymphomyeloid origin of MMCs including DCs and monocytes. Color coding of progenitor populations is according to their expression of CD38 and CD45RA (inset). B/NK, B- and NK-cell progenitor; EMP, erythromyeloid progenitor; EoBa, eosinophil-basophil progenitor; ETP, early thymic progenitor; GM(D)P, granulocyte-monocyte (DC) progenitor; MEP, megaerythroid progenitor; MLP, multi-lymphoid progenitor; MDP, monocyte-DC progenitor.

CD123hi population, but the statement that these cells are “DC restricted” should be qualified to the specified conditions. Under these conditions, GMPs generated both monocytes and DCs. However, strong monocytic differentiation with M-CSF was not tested, and it might therefore be argued that monocyte potential was not completely excluded from the CD123hi fraction. It was also previously shown that CD38+CD10+ lymphoid progenitors have the ability to develop into HLA-DR+CD1a+ DC-like cells and pDCs (100–102). More-recent studies suggest that the CD38–CD45RA+ LMPPs (or multi-lymphoid progenitors), which can give rise to monocytes, will also contain genuine DC potential (85). Whether these lymphoid pathways funnel into the CD123hi population is unknown. Also, it is possible that accessory pathways of DC differentiation exist, as the authors who first described pDC potential in the CD123hi population also noted LC-like potential in the

CD123lo GMP fraction (112); this may indicate the presence of CD1c+ cDC2s (65, 86). The same authors who defined a CDP also reported pre-cDCs in human blood by scrutinizing small populations of CD34–, lineage-negative cells expressing Flt3, GM-CSF, and M-CSF receptors (83). The pre-cDCs that were defined did not express high CD123 and had no pDC potential. The transition between CD123hi CDP and CD123lo pre-cDC is not immediately obvious, and other transitional CD34–, lineage-negative intermediates may exist. Bone marrow transplantation and human DC deficiency states indicate that continual replenishment from blood-borne precursors is required for tissue populations of CD141+ and CD1c+ DCs (7, 43, 44). Comparison with the mouse suggests that these cells are derived from rare pre-DC populations although the potential precursor role of blood DCs has not been excluded (114).

10

ASMscience.org/MicrobiolSpectrum Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Monocyte, Macrophage, and Dendritic Cell Development

MONOCYTE-DERIVED CELLS IN INFLAMMATION The derivation of macrophages from monocytes in vitro has been studied for many years and was one of the founding observations leading to the MPS model. A “classical” highly immune-activated macrophage phenotype is seen after exposure to gamma interferon, while “alternative” activation with IL-4 was developed as a model of resident macrophage differentiation (3). These two phenotypes, known as M1 and M2, respectively, have been regarded as two poles of macrophage differentiation with contrasting functions. More-recent studies have highlighted the complexity of monocyte activation using a greater range of stimuli and employing transcriptomic analysis to derive modules of coregulated genes and their regulators (29). The isolation of primary human macrophages from different sites will now facilitate a more systematic comparison of ex vivo populations with in vitro-derived cells (7, 29, 115, 116). The observation that monocytes could differentiate into myeloid DCs with the ability to stimulate naive T cells was a significant experimental breakthrough facilitating a great number of studies in humans (2). However, recognition of naturally occurring DCs in vivo has led to a reappraisal of the monocyte-derived DC as probably more closely linked with inflammatory monocyte function than to steady-state DC biology. Latterly, the importance of monocyte-derived DCs in resistance to infection has been fully recognized in the mouse, thus validating the human in vitro data (49, 50), and inflammatory DCs have been described in humans in several pathological settings (115–118). In addition to monocyte-derived DCs, the direct recruitment of DC-lineage cells to inflammatory infiltrates remains to be investigated. The problem of separating monocyte-origin and DC-derived inflammatory cells is highlighted by in vitro studies showing that inflammation drives convergent transcriptional programs probably leading to very similar phenotypes (51). Indicators of recent monocyte origin include S100A8/9, recognized by antibody MAC387, a marker long used to describe “monocyte-macrophages” by pathologists (119). Further information is also required to understand the repopulation of LCs following an inflammatory insult. In humans, LCs are replaced by bone marrow-derived cells after HSC transplantation and graft-versus-host disease (79, 80). A two-phase kinetic of LC recovery was observed many years ago in humans in serial skin biopsies of delayed-type hypersensitivity reactions (120). In mice, more detailed analysis shows an initial infiltra-

tion by classical monocytes that express low levels of Langerin (121) followed by a second wave of more longlived LC precursors (122, 123). Langerin+ cells may be derived from monocytes and CD34+ progenitors in vitro (124–126). Recent experiments indicate that monocytes express only a low level of Langerin in comparison with CD1c+ DCs, which rapidly form LC-like cells with Birbeck granules (65, 86). Together, these results suggest that the DC differentiation pathway may contribute to long-term LC repopulation following inflammation.

GENETIC CONTROL OF THE HUMAN MONOCYTE AND DC LINEAGES The genetic control of human monocyte and DC development has been illuminated in a number of ways, using defined growth factors, knockdown of specific genes, and xenograft models (56, 102). Humans with genetic defects of monocyte and DC development have also been described (reviewed in reference 127). DC production from human progenitors is promoted by Flt3 ligand, GM-CSF, IL-4, and the presence of stromal cells (82, 84). Administration of Flt3 ligand to humans expands DCs in vivo (83). GM-CSF is implicated in DC development in mice. This function has not been thoroughly explored in humans, but blockade of GM-CSF signaling by spontaneously occurring autoantibodies (or rare receptor mutations) dramatically inhibits alveolar macrophage development, causing pulmonary alveolar proteinosis (128). Human monocyte and DC development becomes severely impaired in humans with heterozygous GATA2 mutation (129). GATA2 is required for stem cell selfrenewal, and the hemizygous state preferentially affects mononuclear cell development, resulting in a combined DC, monocyte, B, and NK lymphoid (DCML) deficiency (130). The clinical picture of immunodeficiency is variable, but susceptibility to viral infections and mycobacteria is common (131). Flt3 ligand is hugely elevated, probably in response to a fundamental disturbance of stem cell equilibrium rather than as a result of DC deficiency. Ikaros mutation also causes a severe pancytopenia that includes monocytes and probably DCs (132). Neutropenia may involve genes implicated in MMC development in mice, such as GFI1, but monocytes and DCs have not been examined systematically in these disorders (133). Finally, WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome, due to gain-of-function mutation of CXCR4, causes neutropenia and global mononuclear deficiency by preventing leukocytes from leaving the marrow. Clinical

ASMscience.org/MicrobiolSpectrum

11 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Collin and Bigley

manifestations of WHIM may overlap with the features of GATA2 mutation, including warts and susceptibility to mycobacteria (134). Biallelic IRF8 mutation in humans was reported to cause a severe defect of monocytes, myeloid DCs, and pDCs, while a heterozygous state induced only changes in CD1c+ DCs (44). A gene dosage effect may be at work because in the mouse there are substantial differences between the hypomorphic BXH2 mouse, which has a specific defect of cDC1, and the IRF8 gene-deleted mouse, which shows multiple lineage deficiencies and expanded granulopoiesis. Monocytopenia appears to be a direct consequence of the IRF8 dependency of Kruppel-like factor 4 (KLF4) (135), while granulopoiesis is promoted by the failure of IRF8 to suppress CCAAT/enhancerbinding protein α (C/EBPα) activity (136). IRF8 also associates with IRF4, a transcription factor implicated in the development of CD1c DCs (cDC2) (137). The variable effect of heterozygous IRF8 mutation on CD1c+ DCs may be mediated through an IRF8/IRF4 interaction, but further clarification of this is required. In cooperation with IRF8, the activator protein 1 (AP1) factor BATF3 plays a key role in DC development in mice and in human in vitro culture (56, 138). Within the DC lineage, the balance of basic helix-loop-helix protein E2-2 and its inhibitor ID2 is critical for the specification of pDCs or classical DCs from DC precursors (53). Mutation of E2-2 is the cause of Pitt-Hopkins syndrome, in which abnormal pDC development has been noted (139). The recently described transcription factor ZBTB46 is specifically expressed in myeloid CD1c+ DCs and their murine counterparts (31). Although it is not required for DC development, ZBTB46 is very useful in lineage marking and lineage-specific DC depletion to demonstrate that cDC2s have specific roles in immunity (97, 140). Monocytes do not express ZBTB46 at rest, but it is induced when they differentiate into DCs (97).

CONCLUSIONS AND FUTURE PERSPECTIVES The MPS provided a conceptual framework for understanding the development of MMCs for nearly half a century. It is becoming clear that monocytes are not the only axis of homeostasis and that long-lived macrophages and a specialized DC lineage contribute to tissue leukocyte populations. The mouse has been an indispensable model for the analysis of hematopoiesis and continues to provide many insights into the pathways of human monocyte and DC development. While the study of monocyte-derived populations in vitro has furnished many exciting experimental results, the true physiolog-

ical context of this work remains incompletely understood but is likely to provide significant insights into inflammatory monocyte differentiation. A distinct DC lineage is well described in humans, but more studies are required to understand the nonredundant role of these cells in immunity and how they can be manipulated for therapeutic purposes. In the near future, single-cell genomics will provide a new level of detail in the analysis of the progenitor cells and of the genomic and transcriptional architecture controlling cell fate (reviewed in reference 141). This is likely to revise classical models of hematopoiesis; rather than successive bifurcations in cell potential, the picture that is emerging is that differentiation is a graded process punctuated by many phenotypic states. Continuous phenotypic variables are likely to reflect gradients in cell potential, which may be in equilibrium and appear stochastic at given points. A further addition to the modern armamentarium is the use of deep phenotyping and unbiased hierarchical clustering to assist the identification of related populations (142). Intermediate populations may indeed harbor cells with intermediate potentials. The diversity of MMCs provides an important regulatory dimension in immunity and the study of their ontogeny endures as a priority area of research. Although many difficulties relating to their plasticity arise, the ability of progenitors to remain flexible in their potential is likely to be critical in allowing hematopoiesis and immunity to adapt to environmental challenges. ACKNOWLEDGMENTS The authors are grateful to M. A. Haniffa for critical review of the manuscript. V.B. is funded by Wellcome Intermediate Clinical Fellowship WT088555MA.

REFERENCES 1. van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, Langevoort HL. 1972. The mononuclear phagocyte system: a new classification of macrophages, monocytes, and their precursor cells. Bull World Health Organ 46:845–852. 2. Sallusto F, Lanzavecchia A. 1994. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/ macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α. J Exp Med 179:1109–1118. 3. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, Locati M, Mantovani A, Martinez FO, Mege JL, Mosser DM, Natoli G, Saeij JP, Schultze JL, Shirey KA, Sica A, Suttles J, Udalova I, van Ginderachter JA, Vogel SN, Wynn TA. 2014. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41:14–20. 4. Steinman RM, Lustig DS, Cohn ZA. 1974. Identification of a novel cell type in peripheral lymphoid organs of mice. 3. Functional properties in vivo. J Exp Med 139:1431–1445. 5. Hume DA. 2008. Macrophages as APC and the dendritic cell myth. J Immunol 181:5829–5835.

12

ASMscience.org/MicrobiolSpectrum Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Monocyte, Macrophage, and Dendritic Cell Development

6. Merad M, Sathe P, Helft J, Miller J, Mortha A. 2013. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol 31:563–604. 7. Haniffa M, Ginhoux F, Wang XN, Bigley V, Abel M, Dimmick I, Bullock S, Grisotto M, Booth T, Taub P, Hilkens C, Merad M, Collin M. 2009. Differential rates of replacement of human dermal dendritic cells and macrophages during hematopoietic stem cell transplantation. J Exp Med 206:371–385. 8. Wang XN, McGovern N, Gunawan M, Richardson C, Windebank M, Siah TW, Lim HY, Fink K, Li JL, Ng LG, Ginhoux F, Angeli V, Collin M, Haniffa M. 2014. A three-dimensional atlas of human dermal leukocytes, lymphatics, and blood vessels. J Invest Dermatol 134:965–974. 9. McGovern N, Schlitzer A, Gunawan M, Jardine L, Shin A, Poyner E, Green K, Dickinson R, Wang XN, Low D, Best K, Covins S, Milne P, Pagan S, Aljefri K, Windebank M, Miranda-Saavedra D, Larbi A, Wasan PS, Duan K, Poidinger M, Bigley V, Ginhoux F, Collin M, Haniffa M. 2014. Human dermal CD14+ cells are a transient population of monocytederived macrophages. Immunity 41:465–477. 10. Tamoutounour S, Guilliams M, Montanana Sanchis F, Liu H, Terhorst D, Malosse C, Pollet E, Ardouin L, Luche H, Sanchez C, Dalod M, Malissen B, Henri S. 2013. Origins and functional specialization of macrophages and of conventional and monocyte-derived dendritic cells in mouse skin. Immunity 39:925–938. 11. Watchmaker PB, Lahl K, Lee M, Baumjohann D, Morton J, Kim SJ, Zeng R, Dent A, Ansel KM, Diamond B, Hadeiba H, Butcher EC. 2014. Comparative transcriptional and functional profiling defines conserved programs of intestinal DC differentiation in humans and mice. Nat Immunol 15:98–108. 12. Ginhoux F, Jung S. 2014. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol 14:392–404. 13. Bar-On L, Jung S. 2010. Defining dendritic cells by conditional and constitutive cell ablation. Immunol Rev 234:76–89. 14. Seneschal J, Clark RA, Gehad A, Baecher-Allan CM, Kupper TS. 2012. Human epidermal Langerhans cells maintain immune homeostasis in skin by activating skin resident regulatory T cells. Immunity 36:873– 884. 15. Yona S, Kim KW, Wolf Y, Mildner A, Varol D, Breker M, StraussAyali D, Viukov S, Guilliams M, Misharin A, Hume DA, Perlman H, Malissen B, Zelzer E, Jung S. 2013. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38:79–91. 16. Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, Becker CD, See P, Price J, Lucas D, Greter M, Mortha A, Boyer SW, Forsberg EC, Tanaka M, van Rooijen N, García-Sastre A, Stanley ER, Ginhoux F, Frenette PS, Merad M. 2013. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38:792–804. 17. Hoeffel G, Chen J, Lavin Y, Low D, Almeida FF, See P, Beaudin AE, Lum J, Low I, Forsberg EC, Poidinger M, Zolezzi F, Larbi A, Ng LG, Chan JK, Greter M, Becher B, Samokhvalov IM, Merad M, Ginhoux F. 2015. C-Myb+ erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 42:665–678. 18. Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F, Rodewald HR. 2015. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518:547–551. 19. Jakubzick C, Gautier EL, Gibbings SL, Sojka DK, Schlitzer A, Johnson TE, Ivanov S, Duan Q, Bala S, Condon T, van Rooijen N, Grainger JR, Belkaid Y, Ma’ayan A, Riches DW, Yokoyama WM, Ginhoux F, Henson PM, Randolph GJ. 2013. Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes. Immunity 39:599–610. 20. Bain CC, Mowat AM. 2014. Macrophages in intestinal homeostasis and inflammation. Immunol Rev 260:102–117.

21. Lavine KJ, Epelman S, Uchida K, Weber KJ, Nichols CG, Schilling JD, Ornitz DM, Randolph GJ, Mann DL. 2014. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl Acad Sci U S A 111:16029–16034. 22. Massberg S, Schaerli P, Knezevic-Maramica I, Köllnberger M, Tubo N, Moseman EA, Huff IV, Junt T, Wagers AJ, Mazo IB, von Andrian UH. 2007. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell 131:994–1008. 23. Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck DW, Schmitz J. 2000. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol 165:6037–6046. 24. MacDonald KP, Munster DJ, Clark GJ, Dzionek A, Schmitz J, Hart DN. 2002. Characterization of human blood dendritic cell subsets. Blood 100:4512–4520. 25. Miller JC, Brown BD, Shay T, Gautier EL, Jojic V, Cohain A, Pandey G, Leboeuf M, Elpek KG, Helft J, Hashimoto D, Chow A, Price J, Greter M, Bogunovic M, Bellemare-Pelletier A, Frenette PS, Randolph GJ, Turley SJ, Merad M, Immunological Genome Consortium. 2012. Deciphering the transcriptional network of the dendritic cell lineage. Nat Immunol 13:888–899. 26. Gautier EL, Shay T, Miller J, Greter M, Jakubzick C, Ivanov S, Helft J, Chow A, Elpek KG, Gordonov S, Mazloom AR, Ma’ayan A, Chua WJ, Hansen TH, Turley SJ, Merad M, Randolph GJ; Immunological Genome Consortium. 2012. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat Immunol 13:1118–1128. 27. Lavin Y, Winter D, Blecher-Gonen R, David E, Keren-Shaul H, Merad M, Jung S, Amit I. 2014. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159:1312–1326. 28. Guilliams M, Ginhoux F, Jakubzick C, Naik SH, Onai N, Schraml BU, Segura E, Tussiwand R, Yona S. 2014. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol 14:571–578. 29. Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, De Nardo D, Gohel TD, Emde M, Schmidleithner L, Ganesan H, NinoCastro A, Mallmann MR, Labzin L, Theis H, Kraut M, Beyer M, Latz E, Freeman TC, Ulas T, Schultze JL. 2014. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 40:274–288. 30. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, Leenen PJ, Liu YJ, MacPherson G, Randolph GJ, Scherberich J, Schmitz J, Shortman K, Sozzani S, Strobl H, Zembala M, Austyn JM, Lutz MB. 2010. Nomenclature of monocytes and dendritic cells in blood. Blood 116:e74–e80. doi:10.1182/blood-2010-02-258558. 31. Robbins SH, Walzer T, Dembélé D, Thibault C, Defays A, Bessou G, Xu H, Vivier E, Sellars M, Pierre P, Sharp FR, Chan S, Kastner P, Dalod M. 2008. Novel insights into the relationships between dendritic cell subsets in human and mouse revealed by genome-wide expression profiling. Genome Biol 9:R17. doi:10.1186/gb-2008-9-1-r17. 32. Hänsel A, Günther C, Ingwersen J, Starke J, Schmitz M, Bachmann M, Meurer M, Rieber EP, Schäkel K. 2011. Human slan (6-sulfo LacNAc) dendritic cells are inflammatory dermal dendritic cells in psoriasis and drive strong TH17/TH1 T-cell responses. J Allergy Clin Immunol 127:787– 794.e9. doi:10.1016/j.jaci.2010.12.009. 33. Cros J, Cagnard N, Woollard K, Patey N, Zhang SY, Senechal B, Puel A, Biswas SK, Moshous D, Picard C, Jais JP, D’Cruz D, Casanova JL, Trouillet C, Geissmann F. 2010. Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity 33:375–386. 34. Carlin LM, Stamatiades EG, Auffray C, Hanna RN, Glover L, VizcayBarrena G, Hedrick CC, Cook HT, Diebold S, Geissmann F. 2013. Nr4a1-dependent Ly6Clow monocytes monitor endothelial cells and orchestrate their disposal. Cell 153:362–375.

ASMscience.org/MicrobiolSpectrum

13 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Collin and Bigley

35. Hettinger J, Richards DM, Hansson J, Barra MM, Joschko AC, Krijgsveld J, Feuerer M. 2013. Origin of monocytes and macrophages in a committed progenitor. Nat Immunol 14:821–830. 36. Hanna RN, Carlin LM, Hubbeling HG, Nackiewicz D, Green AM, Punt JA, Geissmann F, Hedrick CC. 2011. The transcription factor NR4A1 (Nur77) controls bone marrow differentiation and the survival of Ly6C– monocytes. Nat Immunol 12:778–785. 37. Schmidl C, Renner K, Peter K, Eder R, Lassmann T, Balwierz PJ, Itoh M, Nagao-Sato S, Kawaji H, Carninci P, Suzuki H, Hayashizaki Y, Andreesen R, Hume DA, Hoffmann P, Forrest AR, Kreutz MP, Edinger M, Rehli M, FANTOM consortium. 2014. Transcription and enhancer profiling in human monocyte subsets. Blood 123:e90–e99. doi:10.1182 /blood-2013-02-484188. 38. Abeles RD, McPhail MJ, Sowter D, Antoniades CG, Vergis N, Vijay GK, Xystrakis E, Khamri W, Shawcross DL, Ma Y, Wendon JA, Vergani D. 2012. CD14, CD16 and HLA-DR reliably identifies human monocytes and their subsets in the context of pathologically reduced HLA-DR expression by CD14hi/CD16neg monocytes: expansion of CD14hi/CD16pos and contraction of CD14lo/CD16pos monocytes in acute liver failure. Cytometry A 81:823–834. 39. Fingerle-Rowson G, Angstwurm M, Andreesen R, Ziegler-Heitbrock HW. 1998. Selective depletion of CD14+ CD16+ monocytes by glucocorticoid therapy. Clin Exp Immunol 112:501–506. 40. Damuzzo V, Pinton L, Desantis G, Solito S, Marigo I, Bronte V, Mandruzzato S. 2015. Complexity and challenges in defining myeloidderived suppressor cells. Cytometry B Clin Cytom 88:77–91. 41. Wong KL, Yeap WH, Tai JJ, Ong SM, Dang TM, Wong SC. 2012. The three human monocyte subsets: implications for health and disease. Immunol Res 53:41–57. 42. Cogle CR, Yachnis AT, Laywell ED, Zander DS, Wingard JR, Steindler DA, Scott EW. 2004. Bone marrow transdifferentiation in brain after transplantation: a retrospective study. Lancet 363:1432–1437. 43. Bigley V, Haniffa M, Doulatov S, Wang XN, Dickinson R, McGovern N, Jardine L, Pagan S, Dimmick I, Chua I, Wallis J, Lordan J, Morgan C, Kumararatne DS, Doffinger R, van der Burg M, van Dongen J, Cant A, Dick JE, Hambleton S, Collin M. 2011. The human syndrome of dendritic cell, monocyte, B and NK lymphoid deficiency. J Exp Med 208:227–234. 44. Hambleton S, Salem S, Bustamante J, Bigley V, Boisson-Dupuis S, Azevedo J, Fortin A, Haniffa M, Ceron-Gutierrez L, Bacon CM, Menon G, Trouillet C, McDonald D, Carey P, Ginhoux F, Alsina L, Zumwalt TJ, Kong XF, Kumararatne D, Butler K, Hubeau M, Feinberg J, Al-Muhsen S, Cant A, Abel L, Chaussabel D, Doffinger R, Talesnik E, Grumach A, Duarte A, Abarca K, Moraes-Vasconcelos D, Burk D, Berghuis A, Geissmann F, Collin M, Casanova JL, Gros P. 2011. IRF8 mutations and human dendritic-cell immunodeficiency. N Engl J Med 365:127–138.

stimulation fully differentiates monocytes to DC-SIGN/CD209+ dendritic cells for immune T cell areas. Cell 143:416–429. 50. León B, López-Bravo M, Ardavín C. 2007. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania. Immunity 26:519–531. 51. Manh TP, Alexandre Y, Baranek T, Crozat K, Dalod M. 2013. Plasmacytoid, conventional, and monocyte-derived dendritic cells undergo a profound and convergent genetic reprogramming during their maturation. Eur J Immunol 43:1706–1715. 52. Scott CL, Henri S, Guilliams M. 2014. Mononuclear phagocytes of the intestine, the skin, and the lung. Immunol Rev 262:9–24. 53. Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V. 2011. Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol 29:163–183. 54. Jardine L, Barge D, Ames-Draycott A, Pagan S, Cookson S, Spickett G, Haniffa M, Collin M, Bigley V. 2013. Rapid detection of dendritic cell and monocyte disorders using CD4 as a lineage marker of the human peripheral blood antigen-presenting cell compartment. Front Immunol 4:495. doi:10.3389/fimmu.2013.00495. 55. Crozat K, Guiton R, Contreras V, Feuillet V, Dutertre CA, Ventre E, Vu Manh TP, Baranek T, Storset AK, Marvel J, Boudinot P, Hosmalin A, Schwartz-Cornil I, Dalod M. 2010. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8α+ dendritic cells. J Exp Med 207:1283–1292. 56. Poulin LF, Reyal Y, Uronen-Hansson H, Schraml BU, Sancho D, Murphy KM, Håkansson UK, Moita LF, Agace WW, Bonnet D, Reis e Sousa C. 2012. DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and nonlymphoid tissues. Blood 119:6052–6062. 57. Gurka S, Hartung E, Becker M, Kroczek RA. 2015. Mouse conventional dendritic cells can be universally classified based on the mutually exclusive expression of XCR1 and SIRPα. Front Immunol 6:35. doi:10.3389/fimmu .2015.00035. 58. van de Ven R, van den Hout MF, Lindenberg JJ, Sluijter BJ, van Leeuwen PA, Lougheed SM, Meijer S, van den Tol MP, Scheper RJ, de Gruijl TD. 2011. Characterization of four conventional dendritic cell subsets in human skin-draining lymph nodes in relation to T-cell activation. Blood 118:2502–2510. 59. Mittag D, Proietto AI, Loudovaris T, Mannering SI, Vremec D, Shortman K, Wu L, Harrison LC. 2011. Human dendritic cell subsets from spleen and blood are similar in phenotype and function but modified by donor health status. J Immunol 186:6207–6217.

45. Zaba LC, Fuentes-Duculan J, Steinman RM, Krueger JG, Lowes MA. 2007. Normal human dermis contains distinct populations of CD11c + BDCA-1+ dendritic cells and CD163+FXIIIA+ macrophages. J Clin Invest 117:2517–2525.

60. Haniffa M, Shin A, Bigley V, McGovern N, Teo P, See P, Wasan PS, Wang XN, Malinarich F, Malleret B, Larbi A, Tan P, Zhao H, Poidinger M, Pagan S, Cookson S, Dickinson R, Dimmick I, Jarrett RF, Renia L, Tam J, Song C, Connolly J, Chan JK, Gehring A, Bertoletti A, Collin M, Ginhoux F. 2012. Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103+ nonlymphoid dendritic cells. Immunity 37:60–73.

46. Lenz A, Heine M, Schuler G, Romani N. 1993. Human and murine dermis contain dendritic cells. Isolation by means of a novel method and phenotypical and functional characterization. J Clin Invest 92:2587–2596.

61. Segura E, Durand M, Amigorena S. 2013. Similar antigen crosspresentation capacity and phagocytic functions in all freshly isolated human lymphoid organ-resident dendritic cells. J Exp Med 210:1035–1047.

47. Chu CC, Ali N, Karagiannis P, Di Meglio P, Skowera A, Napolitano L, Barinaga G, Grys K, Sharif-Paghaleh E, Karagiannis SN, Peakman M, Lombardi G, Nestle FO. 2012. Resident CD141 (BDCA3)+ dendritic cells in human skin produce IL-10 and induce regulatory T cells that suppress skin inflammation. J Exp Med 209:935–945.

62. Merad M, Ginhoux F, Collin M. 2008. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol 8:935–947.

48. Jenkins SJ, Ruckerl D, Cook PC, Jones LH, Finkelman FD, van Rooijen N, MacDonald AS, Allen JE. 2011. Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science 332:1284–1288. 49. Cheong C, Matos I, Choi JH, Dandamudi DB, Shrestha E, Longhi MP, Jeffrey KL, Anthony RM, Kluger C, Nchinda G, Koh H, Rodriguez A, Idoyaga J, Pack M, Velinzon K, Park CG, Steinman RM. 2010. Microbial

63. Valladeau J, Ravel O, Dezutter-Dambuyant C, Moore K, Kleijmeer M, Liu Y, Duvert-Frances V, Vincent C, Schmitt D, Davoust J, Caux C, Lebecque S, Saeland S. 2000. Langerin, a novel C-type lectin specific to Langerhans cells, is an endocytic receptor that induces the formation of Birbeck granules. Immunity 12:71–81. 64. Bigley V, McGovern N, Milne P, Dickinson R, Pagan S, Cookson S, Haniffa M, Collin M. 2015. Langerin-expressing dendritic cells in human tissues are related to CD1c+ dendritic cells and distinct from Langerhans cells and CD141high XCR1+ dendritic cells. J Leukoc Biol 97:627–634.

14

ASMscience.org/MicrobiolSpectrum Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Monocyte, Macrophage, and Dendritic Cell Development 65. Milne P, Bigley V, Gunawan M, Haniffa M, Collin M. 2015. CD1c+ blood dendritic cells have Langerhans cell potential. Blood 125:470–473. 66. Romani N, Clausen BE, Stoitzner P. 2010. Langerhans cells and more: langerin-expressing dendritic cell subsets in the skin. Immunol Rev 234: 120–141. 67. Hunger RE, Sieling PA, Ochoa MT, Sugaya M, Burdick AE, Rea TH, Brennan PJ, Belisle JT, Blauvelt A, Porcelli SA, Modlin RL. 2004. Langerhans cells utilize CD1a and langerin to efficiently present nonpeptide antigens to T cells. J Clin Invest 113:701–708. 68. de Witte L, Nabatov A, Pion M, Fluitsma D, de Jong MA, de Gruijl T, Piguet V, van Kooyk Y, Geijtenbeek TB. 2007. Langerin is a natural barrier to HIV-1 transmission by Langerhans cells. Nat Med 13:367–371. 69. Igyártó BZ, Haley K, Ortner D, Bobr A, Gerami-Nejad M, Edelson BT, Zurawski SM, Malissen B, Zurawski G, Berman J, Kaplan DH. 2011. Skin-resident murine dendritic cell subsets promote distinct and opposing antigen-specific T helper cell responses. Immunity 35:260–272. 70. Bennett CL, Fallah-Arani F, Conlan T, Trouillet C, Goold H, Chorro L, Flutter B, Means TK, Geissmann F, Chakraverty R. 2011. Langerhans cells regulate cutaneous injury by licensing CD8 effector cells recruited to the skin. Blood 117:7063–7069. 71. Schuster C, Vaculik C, Fiala C, Meindl S, Brandt O, Imhof M, Stingl G, Eppel W, Elbe-Bürger A. 2009. HLA-DR+ leukocytes acquire CD1 antigens in embryonic and fetal human skin and contain functional antigen-presenting cells. J Exp Med 206:169–181. 72. Hoeffel G, Wang Y, Greter M, See P, Teo P, Malleret B, Leboeuf M, Low D, Oller G, Almeida F, Choy SH, Grisotto M, Renia L, Conway SJ, Stanley ER, Chan JK, Ng LG, Samokhvalov IM, Merad M, Ginhoux F. 2012. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J Exp Med 209:1167–1181. 73. Wang Y, Szretter KJ, Vermi W, Gilfillan S, Rossini C, Cella M, Barrow AD, Diamond MS, Colonna M. 2012. IL-34 is a tissue-restricted ligand of CSF1R required for the development of Langerhans cells and microglia. Nat Immunol 13:753–760. 74. Greter M, Lelios I, Pelczar P, Hoeffel G, Price J, Leboeuf M, Kündig TM, Frei K, Ginhoux F, Merad M, Becher B. 2012. Stroma-derived interleukin-34 controls the development and maintenance of Langerhans cells and the maintenance of microglia. Immunity 37:1050–1060. 75. Borkowski TA, Letterio JJ, Farr AG, Udey MC. 1996. A role for endogenous transforming growth factor β1 in Langerhans cell biology: the skin of transforming growth factor β1 null mice is devoid of epidermal Langerhans cells. J Exp Med 184:2417–2422. 76. Czernielewski JM, Demarchez M. 1987. Further evidence for the self-reproducing capacity of Langerhans cells in human skin. J Invest Dermatol 88:17–20. 77. Merad M, Manz MG, Karsunky H, Wagers A, Peters W, Charo I, Weissman IL, Cyster JG, Engleman EG. 2002. Langerhans cells renew in the skin throughout life under steady-state conditions. Nat Immunol 3:1135–1141. 78. Kanitakis J, Morelon E, Petruzzo P, Badet L, Dubernard JM. 2011. Self-renewal capacity of human epidermal Langerhans cells: observations made on a composite tissue allograft. Exp Dermatol 20:145–146. 79. Collin MP, Hart DN, Jackson GH, Cook G, Cavet J, Mackinnon S, Middleton PG, Dickinson AM. 2006. The fate of human Langerhans cells in hematopoietic stem cell transplantation. J Exp Med 203:27–33. 80. Mielcarek M, Kirkorian AY, Hackman RC, Price J, Storer BE, Wood BL, Leboeuf M, Bogunovic M, Storb R, Inamoto Y, Flowers ME, Martin PJ, Collin M, Merad M. 2014. Langerhans cell homeostasis and turnover after nonmyeloablative and myeloablative allogeneic hematopoietic cell transplantation. Transplantation 98:563–568. 81. Doulatov S, Notta F, Laurenti E, Dick JE. 2012. Hematopoiesis: a human perspective. Cell Stem Cell 10:120–136. 82. Lee J, Breton G, Oliveira TY, Zhou YJ, Aljoufi A, Puhr S, Cameron MJ, Sékaly RP, Nussenzweig MC, Liu K. 2015. Restricted dendritic cell

and monocyte progenitors in human cord blood and bone marrow. J Exp Med 212:385–399. 83. Breton G, Lee J, Zhou YJ, Schreiber JJ, Keler T, Puhr S, Anandasabapathy N, Schlesinger S, Caskey M, Liu K, Nussenzweig MC. 2015. Circulating precursors of human CD1c+ and CD141+ dendritic cells. J Exp Med 212:401–413. 84. Balan S, Ollion V, Colletti N, Chelbi R, Montanana-Sanchis F, Liu H, Vu Manh TP, Sanchez C, Savoret J, Perrot I, Doffin AC, Fossum E, Bechlian D, Chabannon C, Bogen B, Asselin-Paturel C, Shaw M, Soos T, Caux C, Valladeau-Guilemond J, Dalod M. 2014. Human XCR1+ dendritic cells derived in vitro from CD34+ progenitors closely resemble blood dendritic cells, including their adjuvant responsiveness, contrary to monocyte-derived dendritic cells. J Immunol 193:1622–1635. 85. Doulatov S, Notta F, Eppert K, Nguyen LT, Ohashi PS, Dick JE. 2010. Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development. Nat Immunol 11:585–593. 86. Martínez-Cingolani C, Grandclaudon M, Jeanmougin M, Jouve M, Zollinger R, Soumelis V. 2014. Human blood BDCA-1 dendritic cells differentiate into Langerhans-like cells with thymic stromal lymphopoietin and TGF-β. Blood 124:2411–2420. 87. Fogg DK, Sibon C, Miled C, Jung S, Aucouturier P, Littman DR, Cumano A, Geissmann F. 2006. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science 311:83–87. 88. Waskow C, Liu K, Darrasse-Jèze G, Guermonprez P, Ginhoux F, Merad M, Shengelia T, Yao K, Nussenzweig M. 2008. The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues. Nat Immunol 9:676–683. 89. Naik SH, Sathe P, Park HY, Metcalf D, Proietto AI, Dakic A, Carotta S, O’Keeffe M, Bahlo M, Papenfuss A, Kwak JY, Wu L, Shortman K. 2007. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat Immunol 8:1217–1226. 90. Onai N, Obata-Onai A, Schmid MA, Ohteki T, Jarrossay D, Manz MG. 2007. Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat Immunol 8:1207–1216. 91. Naik SH, Metcalf D, van Nieuwenhuijze A, Wicks I, Wu L, O’Keeffe M, Shortman K. 2006. Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes. Nat Immunol 7:663–671. 92. Liu K, Victora GD, Schwickert TA, Guermonprez P, Meredith MM, Yao K, Chu FF, Randolph GJ, Rudensky AY, Nussenzweig M. 2009. In vivo analysis of dendritic cell development and homeostasis. Science 324:392–397. 93. Sathe P, Metcalf D, Vremec D, Naik SH, Langdon WY, Huntington ND, Wu L, Shortman K. 2014. Lymphoid tissue and plasmacytoid dendritic cells and macrophages do not share a common macrophagedendritic cell-restricted progenitor. Immunity 41:104–115. 94. Schraml BU, van Blijswijk J, Zelenay S, Whitney PG, Filby A, Acton SE, Rogers NC, Moncaut N, Carvajal JJ, Reis e Sousa C. 2013. Genetic tracing via DNGR-1 expression history defines dendritic cells as a hematopoietic lineage. Cell 154:843–858. 95. Onai N, Kurabayashi K, Hosoi-Amaike M, Toyama-Sorimachi N, Matsushima K, Inaba K, Ohteki T. 2013. A clonogenic progenitor with prominent plasmacytoid dendritic cell developmental potential. Immunity 38:943–957. 96. Schlitzer A, Heiseke AF, Einwächter H, Reindl W, Schiemann M, Manta CP, See P, Niess JH, Suter T, Ginhoux F, Krug AB. 2012. Tissuespecific differentiation of a circulating CCR9– pDC-like common dendritic cell precursor. Blood 119:6063–6071. 97. Satpathy AT, Kc W, Albring JC, Edelson BT, Kretzer NM, Bhattacharya D, Murphy TL, Murphy KM. 2012. Zbtb46 expression distinguishes classical dendritic cells and their committed progenitors from other immune lineages. J Exp Med 209:1135–1152.

ASMscience.org/MicrobiolSpectrum

15 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Collin and Bigley

98. Sanjuan-Pla A, Macaulay IC, Jensen CT, Woll PS, Luis TC, Mead A, Moore S, Carella C, Matsuoka S, Bouriez Jones T, Chowdhury O, Stenson L, Lutteropp M, Green JC, Facchini R, Boukarabila H, Grover A, Gambardella A, Thongjuea S, Carrelha J, Tarrant P, Atkinson D, Clark SA, Nerlov C, Jacobsen SE. 2013. Platelet-biased stem cells reside at the apex of the haematopoietic stem-cell hierarchy. Nature 502:232–236. 99. Naik SH, Perié L, Swart E, Gerlach C, van Rooij N, de Boer RJ, Schumacher TN. 2013. Diverse and heritable lineage imprinting of early haematopoietic progenitors. Nature 496:229–232. 100. Galy A, Travis M, Cen D, Chen B, Human T. 1995. Human T, B, natural killer, and dendritic cells arise from a common bone marrow progenitor cell subset. Immunity 3:459–473. 101. Chicha L, Jarrossay D, Manz MG. 2004. Clonal type I interferonproducing and dendritic cell precursors are contained in both human lymphoid and myeloid progenitor populations. J Exp Med 200:1519– 1524. 102. Spits H, Couwenberg F, Bakker AQ, Weijer K, Uittenbogaart CH. 2000. Id2 and Id3 inhibit development of CD34+ stem cells into predendritic cell (pre-DC)2 but not into pre-DC1: evidence for a lymphoid origin of pre-DC2. J Exp Med 192:1775–1784. 103. Sathe P, Vremec D, Wu L, Corcoran L, Shortman K. 2013. Convergent differentiation: myeloid and lymphoid pathways to murine plasmacytoid dendritic cells. Blood 121:11–19. 104. Kondo M, Weissman IL, Akashi K. 1997. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 91:661–672. 105. Akashi K, Traver D, Miyamoto T, Weissman IL. 2000. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404:193–197. 106. Kawamoto H, Ikawa T, Masuda K, Wada H, Katsura Y. 2010. A map for lineage restriction of progenitors during hematopoiesis: the essence of the myeloid-based model. Immunol Rev 238:23–36. 107. Goardon N, Marchi E, Atzberger A, Quek L, Schuh A, Soneji S, Woll P, Mead A, Alford KA, Rout R, Chaudhury S, Gilkes A, Knapper S, Beldjord K, Begum S, Rose S, Geddes N, Griffiths M, Standen G, Sternberg A, Cavenagh J, Hunter H, Bowen D, Killick S, Robinson L, Price A, Macintyre E, Virgo P, Burnett A, Craddock C, Enver T, Jacobsen SE, Porcher C, Vyas P. 2011. Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia. Cancer Cell 19:138–152. 108. Ji H, Ehrlich LI, Seita J, Murakami P, Doi A, Lindau P, Lee H, Aryee MJ, Irizarry RA, Kim K, Rossi DJ, Inlay MA, Serwold T, Karsunky H, Ho L, Daley GQ, Weissman IL, Feinberg AP. 2010. Comprehensive methylome map of lineage commitment from haematopoietic progenitors. Nature 467:338–342. 109. Novershtern N, Subramanian A, Lawton LN, Mak RH, Haining WN, McConkey ME, Habib N, Yosef N, Chang CY, Shay T, Frampton GM, Drake AC, Leskov I, Nilsson B, Preffer F, Dombkowski D, Evans JW, Liefeld T, Smutko JS, Chen J, Friedman N, Young RA, Golub TR, Regev A, Ebert BL. 2011. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 144:296–309. 110. Metcalf D. 1997. The molecular control of granulocytes and macrophages. Ciba Found Symp 204:40–50, discussion 50–56. 111. Olweus J, Thompson PA, Lund-Johansen F. 1996. Granulocytic and monocytic differentiation of CD34hi cells is associated with distinct changes in the expression of the PU.1-regulated molecules, CD64 and macrophage colony-stimulating factor receptor. Blood 88:3741–3754. 112. Olweus J, BitMansour A, Warnke R, Thompson PA, Carballido J, Picker LJ, Lund-Johansen F. 1997. Dendritic cell ontogeny: a human dendritic cell lineage of myeloid origin. Proc Natl Acad Sci U S A 94: 12551–12556. 113. Manz MG, Miyamoto T, Akashi K, Weissman IL. 2002. Prospective isolation of human clonogenic common myeloid progenitors. Proc Natl Acad Sci U S A 99:11872–11877. 114. Collin M, McGovern N, Haniffa M. 2013. Human dendritic cell subsets. Immunology 140:22–30.

115. Beitnes AC, Ráki M, Brottveit M, Lundin KE, Jahnsen FL, Sollid LM. 2012. Rapid accumulation of CD14+CD11c+ dendritic cells in gut mucosa of celiac disease after in vivo gluten challenge. PLoS One 7: e33556. doi:10.1371/journal.pone.0033556. 116. Segura E, Touzot M, Bohineust A, Cappuccio A, Chiocchia G, Hosmalin A, Dalod M, Soumelis V, Amigorena S. 2013. Human inflammatory dendritic cells induce Th17 cell differentiation. Immunity 38:336– 348. 117. Wollenberg A, Mommaas M, Oppel T, Schottdorf EM, Günther S, Moderer M. 2002. Expression and function of the mannose receptor CD206 on epidermal dendritic cells in inflammatory skin diseases. J Invest Dermatol 118:327–334. 118. Lowes MA, Chamian F, Abello MV, Fuentes-Duculan J, Lin SL, Nussbaum R, Novitskaya I, Carbonaro H, Cardinale I, Kikuchi T, Gilleaudeau P, Sullivan-Whalen M, Wittkowski KM, Papp K, Garovoy M, Dummer W, Steinman RM, Krueger JG. 2005. Increase in TNF-α and inducible nitric oxide synthase-expressing dendritic cells in psoriasis and reduction with efalizumab (anti-CD11a). Proc Natl Acad Sci U S A 102: 19057–19062. 119. Soulas C, Conerly C, Kim WK, Burdo TH, Alvarez X, Lackner AA, Williams KC. 2011. Recently infiltrating MAC387+ monocytes/ macrophages a third macrophage population involved in SIV and HIV encephalitic lesion formation. Am J Pathol 178:2121–2135. 120. Kaplan G, Nusrat A, Witmer MD, Nath I, Cohn ZA. 1987. Distribution and turnover of Langerhans cells during delayed immune responses in human skin. J Exp Med 165:763–776. 121. Ginhoux F, Tacke F, Angeli V, Bogunovic M, Loubeau M, Dai XM, Stanley ER, Randolph GJ, Merad M. 2006. Langerhans cells arise from monocytes in vivo. Nat Immunol 7:265–273. 122. Seré K, Baek JH, Ober-Blöbaum J, Müller-Newen G, Tacke F, Yokota Y, Zenke M, Hieronymus T. 2012. Two distinct types of Langerhans cells populate the skin during steady state and inflammation. Immunity 37:905–916. 123. Nagao K, Kobayashi T, Moro K, Ohyama M, Adachi T, Kitashima DY, Ueha S, Horiuchi K, Tanizaki H, Kabashima K, Kubo A, Cho YH, Clausen BE, Matsushima K, Suematsu M, Furtado GC, Lira SA, Farber JM, Udey MC, Amagai M. 2012. Stress-induced production of chemokines by hair follicles regulates the trafficking of dendritic cells in skin. Nat Immunol 13:744–752. 124. Geissmann F, Prost C, Monnet JP, Dy M, Brousse N, Hermine O. 1998. Transforming growth factor β1, in the presence of granulocyte/ macrophage colony-stimulating factor and interleukin 4, induces differentiation of human peripheral blood monocytes into dendritic Langerhans cells. J Exp Med 187:961–966. 125. Caux C, Vanbervliet B, Massacrier C, Dezutter-Dambuyant C, de Saint-Vis B, Jacquet C, Yoneda K, Imamura S, Schmitt D, Banchereau J. 1996. CD34+ hematopoietic progenitors from human cord blood differentiate along two independent dendritic cell pathways in response to GM-CSF+TNFα. J Exp Med 184:695–706. 126. Strunk D, Rappersberger K, Egger C, Strobl H, Krömer E, Elbe A, Maurer D, Stingl G. 1996. Generation of human dendritic cells/Langerhans cells from circulating CD34+ hematopoietic progenitor cells. Blood 87: 1292–1302. 127. Collin M, Bigley V, Haniffa M, Hambleton S. 2011. Human dendritic cell deficiency: the missing ID? Nat Rev Immunol 11:575–583. 128. Sakagami T, Uchida K, Suzuki T, Carey BC, Wood RE, Wert SE, Whitsett JA, Trapnell BC, Luisetti M. 2009. Human GM-CSF autoantibodies and reproduction of pulmonary alveolar proteinosis. N Engl J Med 361:2679–2681. 129. Collin M, Dickinson R, Bigley V. 2015. Haematopoietic and immune defects associated with GATA2 mutation. Br J Haematol 169:173–187. 130. Dickinson RE, Milne P, Jardine L, Zandi S, Swierczek SI, McGovern N, Cookson S, Ferozepurwalla Z, Langridge A, Pagan S, Gennery A, Heiskanen-Kosma T, Hämäläinen S, Seppänen M, Helbert M, Tholouli E,

16

ASMscience.org/MicrobiolSpectrum Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40

Monocyte, Macrophage, and Dendritic Cell Development

Gambineri E, Reykdal S, Gottfreðsson M, Thaventhiran JE, Morris E, Hirschfield G, Richter AG, Jolles S, Bacon CM, Hambleton S, Haniffa M, Bryceson Y, Allen C, Prchal JT, Dick JE, Bigley V, Collin M. 2014. The evolution of cellular deficiency in GATA2 mutation. Blood 123:863–874. 131. Spinner MA, Sanchez LA, Hsu AP, Shaw PA, Zerbe CS, Calvo KR, Arthur DC, Gu W, Gould CM, Brewer CC, Cowen EW, Freeman AF, Olivier KN, Uzel G, Zelazny AM, Daub JR, Spalding CD, Claypool RJ, Giri NK, Alter BP, Mace EM, Orange JS, Cuellar-Rodriguez J, Hickstein DD, Holland SM. 2014. GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity. Blood 123:809–821. 132. Goldman FD, Gurel Z, Al-Zubeidi D, Fried AJ, Icardi M, Song C, Dovat S. 2012. Congenital pancytopenia and absence of B lymphocytes in a neonate with a mutation in the Ikaros gene. Pediatr Blood Cancer 58:591–597. 133. Boztug K, Klein C. 2011. Genetic etiologies of severe congenital neutropenia. Curr Opin Pediatr 23:21–26. 134. Hernandez PA, Gorlin RJ, Lukens JN, Taniuchi S, Bohinjec J, Francois F, Klotman ME, Diaz GA. 2003. Mutations in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined immunodeficiency disease. Nat Genet 34:70–74. 135. Kurotaki D, Osato N, Nishiyama A, Yamamoto M, Ban T, Sato H, Nakabayashi J, Umehara M, Miyake N, Matsumoto N, Nakazawa M, Ozato K, Tamura T. 2013. Essential role of the IRF8-KLF4 transcription factor cascade in murine monocyte differentiation. Blood 121:1839– 1849. 136. Kurotaki D, Yamamoto M, Nishiyama A, Uno K, Ban T, Ichino M, Sasaki H, Matsunaga S, Yoshinari M, Ryo A, Nakazawa M, Ozato K, Tamura T. 2014. IRF8 inhibits C/EBPα activity to restrain mononuclear phagocyte progenitors from differentiating into neutrophils. Nat Commun 5:4978. doi:10.1038/ncomms5978.

137. Schlitzer A, McGovern N, Teo P, Zelante T, Atarashi K, Low D, Ho AW, See P, Shin A, Wasan PS, Hoeffel G, Malleret B, Heiseke A, Chew S, Jardine L, Purvis HA, Hilkens CM, Tam J, Poidinger M, Stanley ER, Krug AB, Renia L, Sivasankar B, Ng LG, Collin M, Ricciardi-Castagnoli P, Honda K, Haniffa M, Ginhoux F. 2013. IRF4 transcription factordependent CD11b+ dendritic cells in human and mouse control mucosal IL-17 cytokine responses. Immunity 38:970–983. 138. Tussiwand R, Lee WL, Murphy TL, Mashayekhi M, Kc W, Albring JC, Satpathy AT, Rotondo JA, Edelson BT, Kretzer NM, Wu X, Weiss LA, Glasmacher E, Li P, Liao W, Behnke M, Lam SS, Aurthur CT, Leonard WJ, Singh H, Stallings CL, Sibley LD, Schreiber RD, Murphy KM. 2012. Compensatory dendritic cell development mediated by BATFIRF interactions. Nature 490:502–507. 139. Cisse B, Caton ML, Lehner M, Maeda T, Scheu S, Locksley R, Holmberg D, Zweier C, den Hollander NS, Kant SG, Holter W, Rauch A, Zhuang Y, Reizis B. 2008. Transcription factor E2-2 is an essential and specific regulator of plasmacytoid dendritic cell development. Cell 135: 37–48. 140. Meredith MM, Liu K, Darrasse-Jeze G, Kamphorst AO, Schreiber HA, Guermonprez P, Idoyaga J, Cheong C, Yao KH, Niec RE, Nussenzweig MC. 2012. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J Exp Med 209:1153–1165. 141. Jaitin DA, Keren-Shaul H, Elefant N, Amit I. 2015. Each cell counts: hematopoiesis and immunity research in the era of single cell genomics. Semin Immunol 27:67–71. 142. Becher B, Schlitzer A, Chen J, Mair F, Sumatoh HR, Teng KW, Low D, Ruedl C, Riccardi-Castagnoli P, Poidinger M, Greter M, Ginhoux F, Newell EW. 2014. High-dimensional analysis of the murine myeloid cell system. Nat Immunol 15:1181–1189.

ASMscience.org/MicrobiolSpectrum

17 Downloaded from www.asmscience.org by IP: 128.240.225.13 On: Tue, 03 Jul 2018 09:54:40