Skip to main content
Cellular and Molecular Life Sciences: CMLS logoLink to Cellular and Molecular Life Sciences: CMLS
. 2010 May 1;67(16):2685–2693. doi: 10.1007/s00018-010-0375-x

The multiple roles of monocyte subsets in steady state and inflammation

Clinton S Robbins 1, Filip K Swirski 1,
PMCID: PMC11115635  PMID: 20437077

Abstract

Monocytes participate importantly in immunity. Produced in the bone marrow and released into the blood, they circulate in blood or reside in a spleen reservoir before entering tissue and giving rise to macrophages or dendritic cells. Monocytes are more than transitional cells that adapt to a particular tissue environment indiscriminately. Accumulating evidence now indicates that monocytes are heterogeneous in several species and are themselves predetermined for particular function in the steady state and inflammation. Future therapeutics may harness this heterogeneity to target harmful functions while sparing those that are beneficial. Here, we review recent advances on the ontogeny and function of monocytes and their subsets in humans and mice.

Keywords: Monocyte subsets, Inflammation, Hematopoietic system, Macrophage, Dendritic cell

Introduction

Monocytes are large circulating leukocytes of the myeloid lineage that mediate essential functions of innate immunity including phagocytosis and cytokine production [1, 2]. Generated in the bone marrow, monocytes enter blood and circulate briefly before extravasating to tissue. Upon tissue infiltration, they are believed to differentiate to macrophages or dendritic cells. Thus, they have often been regarded as transitional cells whose eventual phenotypes depend on the particular environment in which they accumulate; in vitro, they are impossible to maintain and either differentiate rapidly or die.

Over the last several years, renewed interest in monocytes has revealed them to be more than simply transitional and reactive. Monocytes are functionally heterogeneous in several species. They can be inflammatory and anti-inflammatory, and they need not differentiate to macrophages or dendritic cells in order to participate in immunity. In sum, monocytes are active constituents of the immune system. In this review, we will discuss the major findings that have enriched our understanding of monocyte subset biology. We will highlight the unanswered pressing questions and discuss some of the lingering controversies.

Human monocyte subsets

The blood of a healthy human adult contains approximately 450 monocytes/μl, which amounts to over 2 billion cells in a 5-l volume. Monocytes are the largest of the circulating leukocytes and can be distinguished morphologically by their characteristic kidney or horseshoe-shaped nucleus, although their variable nucleus to cytoplasm ratio renders monocytes sometimes difficult to distinguish from other leukocytes. It has been known for a long time that monocytes exist [3, 4], but, until recently, they have been regarded primarily as transitional cells whose main function is to repopulate various tissue macrophage populations.

Relying on gradient centrifugation and counterflow centrifugation (elutriation), numerous studies from the early 1980s postulated the existence of morphological and functional monocyte heterogeneity. Collectively, this work showed the presence of a dominant, large and dense “classical” subset that expressed the LPS co-receptor CD14, was phagocytic, produced cytokines including IL-1 and colony-stimulating factor (CSF), had high MPO activity, high antibody-dependent cell-mediated cytotoxicity (ADCC), and suppressed antigen-activated lymphocytes. The second, minor, subset was smaller and less dense, less phagocytic, expressed lower levels of CD14 and higher levels of HLA-DR, exhibited greater capacity to present soluble tetanus toxoid (TT) and particulate antigens, and produced high levels of interferon-alpha [512].

A series of studies conducted in the late 1980s and 1990s focussed on defining human monocyte subsets according to their cell surface antigen expression [13]. One line of inquiry differentiated subsets on the basis of FcγR-I (CD64) expression. Two major subsets were identified. The majority of monocytes were CD64+; these were large, phagocytic, and inflammatory. In contrast, CD64 were rare, smaller, had higher capacity for antigen presentation, and had low cytostatic effects against tumors. In general, then, characterization of monocytes according to CD64 expression recapitulated the earlier studies in which monocyte subsets were separated by size and density [1420].

A second line of inquiry focused on the expression of FcγR-III (CD16) [21, 22]. It was shown that the majority of monocytes were CD16, had low expression of HLA-DR, were highly phagocytic and inflammatory, and expressed the chemokine receptor CCR2. A minor subset was CD16+, HLA-DR+, CCR2, and CCR5+. Given the finding that CD16 monocytes can upregulate CD16 and that macrophages express CD16, it was postulated that the CD16+ subset was a more mature version of CD16 monocytes [23]. CD16+ monocytes were termed pro-inflammatory because of their capacity to produce TNF-α in response to LPS [24, 25], and subsequent studies have shown that this subset can differentiate to dendritic cells, respond to fractalkine [2628], and increase under various stress or disease conditions [2933].

Grage-Griebenow and colleagues [13] proposed the existence of at least three subsets of human monocytes, two of them within the CD16+ population. It was shown, for example, that CD16+CD14high monocytes respond differentially to LPS, zymosan, and Staphylococcus aureus compared to CD16+CD14dim monocytes. Whereas CD16+CD14high monocytes are the main producers of IL-10, the CD16+CD14dim monocytes are major producers of TNF-α, but only in response to LPS and not to zymosan or S. aureus [34].

Accumulating evidence nevertheless suggests that it is the CD16 monocyte subset that is more inflammatory: CD16 monocytes express CCR2 and MPO at higher levels [35], phenotypically resemble inflammatory monocytes in mice (see below) [2], rise in the blood shortly after myocardial infarction [36], and upregulate PD-1, an anti-inflammatory co-stimulatory molecule, slightly less than CD16+ monocytes [37]. Perhaps the more important question, then, is not which subset is inflammatory and which is anti-inflammatory, but how a particular subset responds to a given stimulus.

Demarcation of human monocyte subsets according to the expression of CD16 has endured, but, in the absence of correlates in animal models, insight into the functional consequence of monocyte heterogeneity was limited to ex vivo and in vitro approaches. With the discovery of mouse monocyte subsets, however, it became possible to investigate monocyte heterogeneity in vivo.

Mouse monocyte subsets

In contrast to humans, mouse monocytes represent about 1.5% of the total peripheral blood leukocyte pool, which corresponds to fewer than 100 cells/μl blood volume. This rarity, combined with the difficulty in maintaining them in culture, has made it difficult to study monocytes in the mouse. In 2000, Jung et al. [38] reported the generation of a transgenic mouse that expresses GFP under the CX3CR1 promoter. Cells that typically express the chemokine receptor could now be visualized microscopically or by flow cytometry. Phenotypic characterization of either homozygous GFP+ mice (and hence CX3CR1-deficient) or heterozygous GFP+ CX3CR1+ mice revealed that GFP+ cells were mostly monocytes and their tissue descendants. A study by von Andrian and colleagues [39] in 2001 that utilized these mice revealed two CD11bhighF4/80+ populations that differed according to GFP expression: GFPhigh cells were CCR2lowCD62Llow while GFPmed cells were CCR2highCD62Lhigh. The existence of two distinct subsets was postulated, but functional studies were not pursued. In 2003, Geissmann et al. [40] described the phenotype and functional relevance of monocyte subsets. In a series of profiling and fate mapping studies, it was shown that the main subset is large, Ly-6Chigh(Gr-1+) CX3CR1lowCCR2highCD62L+, has a short half-life, and migrates to inflamed tissues. A second subset is smaller in size, Ly-6Clow(Gr-1) CX3CR1highCCR2low/negCD62Lneg and is found in inflamed and resting tissue. Accordingly, Ly-6Chigh monocytes were called ‘inflammatory’ while Ly-6Clow monocytes were termed ‘resident’. Collectively, these studies provided the tools and stimulated further interest into how monocyte subsets uniquely influence the immune response (Fig. 1).

Fig. 1.

Fig. 1

The development, phenotype, differentiation, and distribution of mouse monocyte subsets and allied myeloid cells. Cartoon depicts the development of monocyte subsets in the bone marrow, their entrance into the blood and accumulation in tissue. Brown arrows depict migration and black arrows depict differentiation. Markers listed in brown depict molecules by which movement is mediated. Ly-6C high Ly-6Chigh monocyte, Ly-6C low Ly-6Clow monocyte, GMP granulocyte and macrophage progenitor, MDP macrophage and dendritic cell progenitor, CDP common dendritic cell progenitor, preDC pre-dendritic cell, TipDC TNFα and iNOS-producing dendritic cell, macrophage

The origin of mouse monocytes and their subsets

Monocytes belong to the myeloid lineage and develop along a path of increasing phenotypic restriction and commitment starting with the earliest progenitor, the hematopoietic stem cell (HSC). Downstream, monocyte restriction follows along the common myeloid progenitor (CMP) and the granulocyte and macrophage progenitor (GMP). GMPs can give rise to monocytes and neutrophils but not megakaryocytes, erythrocytes, or lymphocytes [41, 42]. Monocyte commitment depends on growth factors such as M-CSF acting via its ligand M-CSFR, and involves transcription factors such as PU.1, KLF-4, MafB, and c-Maf [1].

Recently, several groups have characterized the developmental pathways of monocytes, macrophages, and dendritic cells downstream of the GMP. Geissmann and colleagues described progenitors that express the fractalkine receptor CX3CR1, c-kit (CD117) [43] and CD115 [44] downstream of the GMP. Termed macrophage and dendritic cell progenitors (MDP), they give rise to monocytes, macrophages, and classical dendritic cell subsets, but not neutrophils. Separate groups have described dedicated dendritic cell precursors, termed common dendritic cell precursors (CDP) that express Flt-3 (CD135) and give rise to classical dendritic cells (both CD8+ and CD8) as well as plasmacytoid dendritic cells (pDC), but not monocytes [45, 46]. Phenotypically, MDPs and CDPs differ according to c-kit expression: MDPs express c-kit at higher levels than CDPs. Fate-mapping experiments have determined that these precursors fall on a continuum of increased phenotypic restriction: CDPs are most restricted because they give rise to DCs at the exclusion of monocytes or macrophages; MDPs give rise to DCs, monocytes and macrophages; GMPs give rise to all these phenotypes as well as to neutrophils [47].

Overall, these studies have been insightful about the nature of monocytes and their relationship to dendritic cells and macrophages. A salient observation is that monocytes do not give rise to classical dendritic cells. Classical dendritic cells (sometimes called “conventional” dendritic cells) were discovered by Steinman and Cohn [4850] more than 30 years ago. They were first shown to reside in secondary lymphoid organs such as the spleen. Today, we know that there are many dendritic cell subsets found in various organs that are specialized for different immune functions [51]. The fact that many of these subsets do not derive from monocytes but from dedicated precursors challenges our assumptions that monocytes’ main role is to repopulate tissue dendritic cells and macrophages.

A lingering ontogeny issue of some contention is the relationship between subsets. Is one subset a more mature version of another or do subsets derive from separate precursors? As mentioned above, it is believed that the CD16+ human monocyte subset is a more mature form of the CD16 subset. Likewise in the mouse, a series of experiments have argued that Ly-6Chigh monocytes convert to Ly-6Clow monocytes. In support of this, Sunderkotter et al. [52] depleted peripheral blood monocytes by clodronate-rich liposomes and showed sequential repopulation of subsets: Ly-6Chigh monocytes appeared in the circulation first; Ly-6Clow monocytes emerged next. Fate-mapping experiments by Varol et al. [53] argued that Ly-6Chigh accumulate in the bone marrow and convert to Ly-6Clow monocytes before re-entering the blood. Given that Ly-6Clow monocytes circulate longer than Ly-6Chigh monocytes [54], it is possible that the sequential appearance of Ly-6Chigh and Ly-6Clow monocytes indicates separate and differently-timed differentiation of either subset rather than conversion. However, a dedicated precursor that gives rise to Ly-6Clow monocytes in the absence of a Ly-6Chigh intermediate has not been identified and therefore conversion remains a plausible mechanism by which the two subsets are linked.

The location where monocytes originate has also received attention lately. It has been believed that monocytes are exclusively generated and stored in the bone marrow. It is a paradigm first proposed in 1968 [4] and supported by many studies, including those investigating monocyte progenitors [1]. It has recently been shown, however, that bona fide monocytes also reside in a splenic reservoir in large numbers [55]. Splenic monocytes cluster in the subcapsular red pulp and are distinct from other macrophage and dendritic cell populations in the spleen. Unlike bone marrow monocytes, which require CCR2 to enter the circulation [56, 57], splenic monocyte exit is independent of CCR2. Signals such as angiotensin II mobilize splenic monocytes en masse, which then enter the circulation and mediate immunity in other tissue sites.

The function of mouse monocyte subsets

Monocyte accumulation in the blood

A growing body of literature indicates that Ly-6Chigh monocytes preferentially accumulate in the circulation during acute and chronic inflammation. Bone marrow-derived Ly-6Chigh monocytes enter the blood via CCR2, a chemokine receptor they express at high levels [5658]. In response to acute injury or infection, the number of Ly-6Chigh monocytes rapidly and dramatically increases in the circulation [59, 60]. This increase is conferred not only by a heightened production and release of monocytes from the bone marrow, but also by their mobilization from a splenic monocyte reservoir [55]. The presence of large numbers of Ly-6Chigh monocytes in the circulation likely confers a stochastic advantage for their eventual accumulation in the affected site. As injury wanes or infection is resolved, Ly-6Chigh monocyte numbers wane in the circulation by mechanisms that are still poorly understood but almost certainly involve a decrease in the rate of cell production. During chronic inflammation in which the inflammatory insult persists, such as atherosclerosis, the number of circulating Ly-6Chigh monocytes increases progressively [61, 62]. Consequently, monocytes themselves contribute to the pathology and, by extension, represent potential therapeutic targets. To what extent Ly-6Clow monocytes increase in the circulation is a matter of some debate. In models of acute inflammation such as myocardial infarction, Ly-6Clow monocyte numbers tend to remain relatively unchanged [60]. In chronic inflammation, such as during atherosclerosis, different groups have observed a range of Ly-6Clow monocyte levels [6265]. It has also been argued that Ly-6Clow monocytes selectively accumulate oxidized lipoproteins, raising the intriguing possibility that, even if less numerous, Ly-6Clow monocytes contribute importantly to atherosclerosis by acting as Trojan horses and bringing lipids to the growing atheroma. Clearly, more research is required to address how monocyte numbers are controlled in the steady state and during inflammation.

Monocyte accumulation in tissue depends on chemokines and their receptors

Accumulation of monocyte subsets into peripheral tissues depends on a myriad of adhesion molecules and chemokines/chemokine receptors. Deficiency of adhesion molecules such as VCAM-1, ICAM-1, or CD18/β2 is known to inhibit atherosclerosis, a disease that depends on monocyte accumulation in the growing atheromata [66]. Chemokines and their receptors have received considerable attention in the context of monocyte subsets because subsets are typically differentiated according to their chemokine receptor expression profile [67, 68]. In experimentally-induced myocardial infarction, CCR2/MCP1 signaling drives the early accumulation of Ly-6Chigh monocytes to the infarcted myocardium while CX3CR1/fractalkine signaling is more important to the later accumulation of Ly-6Clow monocytes [60]. In infection, CCR2-mediated recruitment of Ly-6Chigh monocytes from the bone marrow to the circulation depends on MCP-1 and MCP-3, although CCR2 is not required for the accumulation of these cells to the spleen [69]. CCR6/CCL20 have also been implicated in the accumulation of monocytes to tissue [70]. In models of atherosclerosis, a number of chemokine/chemokine receptor pairs relevant to monocyte recruitment have been implicated and include CCR2/MCP-1, CX3CR1/fractalkine, and CCR5/MIP-1α [71]. Atherosclerosis is markedly reduced when CCR2/MCP-1 and CX3CR1/fractalkine signaling is inhibited, and is almost absent through additional inhibition of CCR5 [63, 7274]. These studies indicate that in atherosclerosis both subsets accumulate to promote disease, despite the observation that Ly-6Chigh monocytes accumulate more frequently [62, 65, 7578]. It is important to note, however, that chemokines and their receptors are not engaged exclusively in chemotaxis. For example, CX3CR1 has been implicated in cell survival [79]. Future studies will need to investigate the contribution of these molecules to such processes as proliferation, differentiation, and survival, in addition to their reported role in mobilization from the bone marrow and accumulation in tissue.

Monocytes are functionally heterogenous upon accumulation

Several groups have shown that, upon accumulation, monocyte subsets and their progeny are functionally heterogeneous. In myocardial infarction, early-accumulating Ly-6Chigh monocytes scavenge necrotic debris and promote inflammation through the production of TNF-α and proteolytic enzymes [60]. Likewise, in response to bacterial, viral, or parasitic infection, Ly-6Chigh monocytes are inflammatory and produce a combination of TNF-α, iNOS, IL-12, and type 1 interferon [8086]. It has also been shown in a model of skeletal muscle injury that Ly-6Chigh monocytes acquire a less inflammatory profile in tissue characterized by reduced Ly-6C expression and upregulation of CX3CR1, CD11c, and IL-10 [87]. Ly-6Clow monocytes, on the other hand, contribute to wound healing in the infarcted myocardium rather than inflammation: they are a source of VEGF and their accumulation coincides with angiogenesis and increased collagen deposition. Ly-6Clow monocytes may also be the first to respond following injury or infection. In the steady state, they crawl along the luminal side of blood vessels and patrol the vasculature [88], a behavior that is probably not unique to monocytes but may be essential to how these cells sense their environment [89, 90]. Within the first couple of hours following infection they produce TNFα transiently [88].

Differentiation potential of monocytes in tissue

Differential accumulation and function of monocyte subsets in tissue may also indicate that monocyte subsets give rise to different progeny. One idea is that monocyte subsets differ in their capacity to give rise to either dendritic cells or macrophages. Given that Ly-6Clow monocytes express CD11c at low levels, and that they upregulate CD11c upon accumulation to atherosclerotic lesions, it was postulated that Ly-6Clow monocytes are precursors of dendritic cells while Ly-6Chigh monocytes preferentially give rise to macrophages. Evaluation of transcription factors such as Pu.1, RelB, cMaf, and Mafb involved in dendritic cell and macrophage differentiation has led some to conclude, however, that Ly-6Chigh monocytes accumulating in the peritoneum are more prone toward dendritic cell differentiation while Ly-6Clow monocytes become macrophages [88, 91]. Pamer and colleagues [82] have described TNFα and iNOS-producing dendritic cells, termed TipDC, that differentiate from Ly-6Chigh monocytes in response to Listeria infection. The process of differentiation is MyD88-dependent [85, 92], and studies now suggest Ly-6Chigh monocyte-derived DCs may be important in cross-priming [70, 93, 94]. Jung and colleagues have explored the differentiation potential of blood monocytes in the lung. They reported that Ly-6Clow monocytes give rise to lung macrophages [95] while Ly-6Chigh monocytes differentiate to dendritic cells in the lung but not in the spleen [53]. The authors further state, however, that Ly-6Chigh monocytes can convert to Ly-6Clow monocytes in the bone marrow and, therefore, also differentiate to macrophages.

A second possibility is that monocyte subsets can be grouped according to the particular macrophage or dendritic cell phenotype they eventually acquire. M1 macrophages, or classically-activated macrophages, arise in vitro when bone marrow-derived cells are cultured with inflammatory triggers such as IFNγ or LPS, whereas M2 macrophages, or alternatively-activated macrophages, arise when cells are cultured with cytokines typically associated with the resolution of inflammation, such as IL-10. M1 macrophages typically express inflammatory mediators such as IL-1, iNOS, ICAM, and MMP9 while M2 macrophages express arginase, CCR7, IL-10, and Fizz, among others. Polarization of these macrophage populations represents an extreme, and there is likely a continuum of phenotypes in vivo [96, 97]. It is possible therefore, that Ly-6Chigh monocytes give rise to M1 macrophages while Ly-6Clow monocytes give rise to M2 macrophages. This is supported by studies in a model of myocardial infarction in which Ly-6Chigh monocytes that accumulated early were inflammatory whereas Ly-6Clow monocytes that accumulated later mediated granulation tissue formation and resolved inflammation, a finding that was consistent in blood of patients with primary acute myocardial infarction [36]. Demarcation of monocyte subsets as M1 and M2 has also been observed in kidney injury [98], and in response to Toxoplasma gondii infection where it was shown that Ly6Chigh monocytes migrated to the infected ileum where they produced TNFα and iNOS, and upregulated F4/80, but not CD11c [83].

Subsets may also be grouped according to the type of DC phenotype they acquire [99]. Randolph and colleagues have shown that Ly-6Chigh monocytes preferentially give rise to CD103+ DCs in the lung whereas Ly-6Clow monocytes give rise to CD103 DCs [100]. In the lamina propria, Jung and colleagues [101] have shown that Ly-6Chigh monocytes preferentially give rise to CD103 DCs while CD103+ DCs arise from MDPs without a monocyte intermediate. In the skin, Merad and colleagues [102] have shown that Langerhans cells may arise from Ly-6Chigh monocytes, although recent studies indicate that the contribution of Ly-6Chigh monocyte-derived Langerhans cells may be less numerous than previously believed [103]. Ly-6Chigh monocytes may also give rise to microglia during inflammation [104]. Finally, studies in tumor-bearing mice reveal that Ly-6Chigh monocytes may be the precursors of myeloid-derived suppressor cells (MDSC) [105108]. Studies are needed, however, to investigate the relationship between monocytes and MDSC in more detail.

Concluding remarks

The discovery that monocyte subset heterogeneity is conserved between species is important to the study of functional differences between monocyte populations in human health and disease. Today, we are, however, learning that human monocyte subsets and mouse monocyte subsets are not fully equivalent [109], and a number of conflicting data and controversies still need to be addressed. Despite this, renewed interest in monocytes over the last several years has revealed some fundamental mechanisms by which these cells participate in immunity. Ly6Chigh and Ly6Clow monocytes function in a highly orchestrated and complementary manner during the development of the immune response. Monocytes are not simply intermediates transitioning to tissue macrophages and dendritic cells, but function as effectors of the immune system. Continued study of functional differences between monocyte subsets will increase our understanding of the underlying processes involved in the initiation and resolution of inflammatory processes.

Acknowledgments

This work was funded by NIH grant 1R01HL095612 (to FKS) and an AHA postdoctoral fellowship (to CSR).

References

  • 1.Auffray C, Sieweke MH, Geissmann F. Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol. 2009;27:669–692. doi: 10.1146/annurev.immunol.021908.132557. [DOI] [PubMed] [Google Scholar]
  • 2.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5:953–964. doi: 10.1038/nri1733. [DOI] [PubMed] [Google Scholar]
  • 3.Sabin FR, Doan CA. The relation of monocytes and clasmatocytes to early infection in rabbits with bovine Tubercle Bacilli. J Exp Med. 1927;46:627–644. doi: 10.1084/jem.46.4.627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.van Furth R, Cohn ZA. The origin and kinetics of mononuclear phagocytes. J Exp Med. 1968;128:415–435. doi: 10.1084/jem.128.3.415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yasaka T, Mantich NM, Boxer LA, Baehner RL. Functions of human monocyte and lymphocyte subsets obtained by countercurrent centrifugal elutriation: differing functional capacities of human monocyte subsets. J Immunol. 1981;127:1515–1518. [PubMed] [Google Scholar]
  • 6.Figdor CG, Bont WS, Touw I, de Roos J, Roosnek EE, de Vries JE. Isolation of functionally different human monocytes by counterflow centrifugation elutriation. Blood. 1982;60:46–53. [PubMed] [Google Scholar]
  • 7.Akiyama Y, Miller PJ, Thurman GB, Neubauer RH, Oliver C, Favilla T, Beman JA, Oldham RK, Stevenson HC. Characterization of a human blood monocyte subset with low peroxidase activity. J Clin Invest. 1983;72:1093–1105. doi: 10.1172/JCI111034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schreiber AD, Kelley M, Dziarski A, Levinson AI. Human monocyte functional heterogeneity: monocyte fractionation by discontinuous albumin gradient centrifugation. Immunology. 1983;49:231–238. [PMC free article] [PubMed] [Google Scholar]
  • 9.Weiner RS, Mason RR. Subfractionation of human blood monocyte subsets with Percoll. Exp Hematol. 1984;12:800–804. [PubMed] [Google Scholar]
  • 10.Akiyama Y, Stevenson GW, Schlick E, Matsushima K, Miller PJ, Stevenson HC. Differential ability of human blood monocyte subsets to release various cytokines. J Leukoc Biol. 1985;37:519–530. doi: 10.1002/jlb.37.5.519. [DOI] [PubMed] [Google Scholar]
  • 11.Elias JA, Chien P, Gustilo KM, Schreiber AD. Differential interleukin-1 elaboration by density-defined human monocyte subpopulations. Blood. 1985;66:298–301. [PubMed] [Google Scholar]
  • 12.Esa AH, Noga SJ, Donnenberg AD, Hess AD. Immunological heterogeneity of human monocyte subsets prepared by counterflow centrifugation elutriation. Immunology. 1986;59:95–99. [PMC free article] [PubMed] [Google Scholar]
  • 13.Grage-Griebenow E, Flad HD, Ernst M. Heterogeneity of human peripheral blood monocyte subsets. J Leukoc Biol. 2001;69:11–20. [PubMed] [Google Scholar]
  • 14.Zembala M, Uracz W, Ruggiero I, Mytar B, Pryjma J. Isolation and functional characteristics of FcR+ and FcR- human monocyte subsets. J Immunol. 1984;133:1293–1299. [PubMed] [Google Scholar]
  • 15.Pryjma J, Pituch-Noworolska A, Ruggiero I, Zembala M. The regulation of polyclonal immunoglobulin synthesis by FcR+ and FcR− monocyte subsets. Clin Immunol Immunopathol. 1985;37:245–252. doi: 10.1016/0090-1229(85)90156-4. [DOI] [PubMed] [Google Scholar]
  • 16.Grage-Griebenow E, Lorenzen D, Fetting R, Flad HD, Ernst M. Phenotypical and functional characterization of Fc gamma receptor I (CD64)-negative monocytes, a minor human monocyte subpopulation with high accessory and antiviral activity. Eur J Immunol. 1993;23:3126–3135. doi: 10.1002/eji.1830231213. [DOI] [PubMed] [Google Scholar]
  • 17.Grage-Griebenow E, Flad HD, Ernst M. Fc gamma receptor I (CD64)-negative human monocytes are potent accessory cells in viral antigen-induced T cell activation and exhibit high IFN-alpha-producing capacity. J Leukoc Biol. 1996;60:389–396. doi: 10.1002/jlb.60.3.389. [DOI] [PubMed] [Google Scholar]
  • 18.Grage-Griebenow E, Baran J, Loppnow H, Los M, Ernst M, Flad HD, Pryjma J. An Fc gamma receptor I (CD64)-negative subpopulation of human peripheral blood monocytes is resistant to killing by antigen-activated CD4-positive cytotoxic T cells. Eur J Immunol. 1997;27:2358–2365. doi: 10.1002/eji.1830270934. [DOI] [PubMed] [Google Scholar]
  • 19.Grage-Griebenow E, Flad HD, Ernst M, Bzowska M, Skrzeczynska J, Pryjma J. Human MO subsets as defined by expression of CD64 and CD16 differ in phagocytic activity and generation of oxygen intermediates. Immunobiology. 2000;202:42–50. doi: 10.1016/S0171-2985(00)80051-0. [DOI] [PubMed] [Google Scholar]
  • 20.Grage-Griebenow E, Zawatzky R, Kahlert H, Brade L, Flad H, Ernst M. Identification of a novel dendritic cell-like subset of CD64(+)/CD16(+) blood monocytes. Eur J Immunol. 2001;31:48–56. doi: 10.1002/1521-4141(200101)31:1<48::AID-IMMU48>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
  • 21.Passlick B, Flieger D, Ziegler-Heitbrock HW. Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood. 1989;74:2527–2534. [PubMed] [Google Scholar]
  • 22.Ziegler-Heitbrock HW, Passlick B, Flieger D. The monoclonal antimonocyte antibody My4 stains B lymphocytes and two distinct monocyte subsets in human peripheral blood. Hybridoma. 1988;7:521–527. doi: 10.1089/hyb.1988.7.521. [DOI] [PubMed] [Google Scholar]
  • 23.Ziegler-Heitbrock HW, Fingerle G, Strobel M, Schraut W, Stelter F, Schutt C, Passlick B, Pforte A. The novel subset of CD14+/CD16+ blood monocytes exhibits features of tissue macrophages. Eur J Immunol. 1993;23:2053–2058. doi: 10.1002/eji.1830230902. [DOI] [PubMed] [Google Scholar]
  • 24.Belge KU, Dayyani F, Horelt A, Siedlar M, Frankenberger M, Frankenberger B, Espevik T, Ziegler-Heitbrock L. The proinflammatory CD14 + CD16 + DR++ monocytes are a major source of TNF. J Immunol. 2002;168:3536–3542. doi: 10.4049/jimmunol.168.7.3536. [DOI] [PubMed] [Google Scholar]
  • 25.Frankenberger M, Sternsdorf T, Pechumer H, Pforte A, Ziegler-Heitbrock HW. Differential cytokine expression in human blood monocyte subpopulations: a polymerase chain reaction analysis. Blood. 1996;87:373–377. [PubMed] [Google Scholar]
  • 26.Sanchez-Torres C, Garcia-Romo GS, Cornejo-Cortes MA, Rivas-Carvalho A, Sanchez-Schmitz G. CD16+ and CD16− human blood monocyte subsets differentiate in vitro to dendritic cells with different abilities to stimulate CD4+ T cells. Int Immunol. 2001;13:1571–1581. doi: 10.1093/intimm/13.12.1571. [DOI] [PubMed] [Google Scholar]
  • 27.Randolph GJ, Sanchez-Schmitz G, Liebman RM, Schakel K. The CD16(+) (FcgammaRIII(+)) subset of human monocytes preferentially becomes migratory dendritic cells in a model tissue setting. J Exp Med. 2002;196:517–527. doi: 10.1084/jem.20011608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ancuta P, Rao R, Moses A, Mehle A, Shaw SK, Luscinskas FW, Gabuzda D. Fractalkine preferentially mediates arrest and migration of CD16+ monocytes. J Exp Med. 2003;197:1701–1707. doi: 10.1084/jem.20022156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ziegler-Heitbrock L. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leukoc Biol. 2007;81:584–592. doi: 10.1189/jlb.0806510. [DOI] [PubMed] [Google Scholar]
  • 30.Horelt A, Belge KU, Steppich B, Prinz J, Ziegler-Heitbrock L. The CD14+ CD16+ monocytes in erysipelas are expanded and show reduced cytokine production. Eur J Immunol. 2002;32:1319–1327. doi: 10.1002/1521-4141(200205)32:5<1319::AID-IMMU1319>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
  • 31.Fingerle-Rowson G, Auers J, Kreuzer E, Fraunberger P, Blumenstein M, Ziegler-Heitbrock LH. Expansion of CD14+ CD16+ monocytes in critically ill cardiac surgery patients. Inflammation. 1998;22:367–379. doi: 10.1023/A:1022316815196. [DOI] [PubMed] [Google Scholar]
  • 32.Mizuno K, Toma T, Tsukiji H, Okamoto H, Yamazaki H, Ohta K, Ohta K, Kasahara Y, Koizumi S, Yachie A. Selective expansion of CD16highCCR2- subpopulation of circulating monocytes with preferential production of haem oxygenase (HO)-1 in response to acute inflammation. Clin Exp Immunol. 2005;142:461–470. doi: 10.1111/j.1365-2249.2005.02932.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Noel JG, Osterburg A, Wang Q, Guo X, Byrum D, Schwemberger S, Goetzman H, Caldwell CC, Ogle CK. Thermal injury elevates the inflammatory monocyte subpopulation in multiple compartments. Shock. 2007;28:684–693. doi: 10.1097/shk.0b013e31805362ed. [DOI] [PubMed] [Google Scholar]
  • 34.Skrzeczynska-Moncznik J, Bzowska M, Loseke S, Grage-Griebenow E, Zembala M, Pryjma J. Peripheral blood CD14 high CD16+ monocytes are main producers of IL-10. Scand J Immunol. 2008;67:152–159. doi: 10.1111/j.1365-3083.2007.02051.x. [DOI] [PubMed] [Google Scholar]
  • 35.Wildgruber M, Lee H, Chudnovskiy A, Yoon TJ, Etzrodt M, Pittet MJ, Nahrendorf M, Croce K, Libby P, Weissleder R, Swirski FK. Monocyte subset dynamics in human atherosclerosis can be profiled with magnetic nano-sensors. PLoS One. 2009;4:e5663. doi: 10.1371/journal.pone.0005663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tsujioka H, Imanishi T, Ikejima H, Kuroi A, Takarada S, Tanimoto T, Kitabata H, Okochi K, Arita Y, Ishibashi K, Komukai K, Kataiwa H, Nakamura N, Hirata K, Tanaka A, Akasaka T. Impact of heterogeneity of human peripheral blood monocyte subsets on myocardial salvage in patients with primary acute myocardial infarction. J Am Coll Cardiol. 2009;54:130–138. doi: 10.1016/j.jacc.2009.04.021. [DOI] [PubMed] [Google Scholar]
  • 37.Said EA, Dupuy FP, Trautmann L, Zhang Y, Shi Y, El-Far M, Hill BJ, Noto A, Ancuta P, Peretz Y, Fonseca SG, Van Grevenynghe J, Boulassel MR, Bruneau J, Shoukry NH, Routy JP, Douek DC, Haddad EK, Sekaly RP. Programmed death-1-induced interleukin-10 production by monocytes impairs CD4(+) T cell activation during HIV infection. Nat Med. 2010;16:452–459. doi: 10.1038/nm.2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, Littman DR. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol. 2000;20:4106–4114. doi: 10.1128/MCB.20.11.4106-4114.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Palframan RT, Jung S, Cheng G, Weninger W, Luo Y, Dorf M, Littman DR, Rollins BJ, Zweerink H, Rot A, von Andrian UH. Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med. 2001;194:1361–1373. doi: 10.1084/jem.194.9.1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003;19:71–82. doi: 10.1016/S1074-7613(03)00174-2. [DOI] [PubMed] [Google Scholar]
  • 41.Akashi K, Traver D, Miyamoto T, Weissman IL. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature. 2000;404:193–197. doi: 10.1038/35004599. [DOI] [PubMed] [Google Scholar]
  • 42.Kondo M, Wagers AJ, Manz MG, Prohaska SS, Scherer DC, Beilhack GF, Shizuru JA, Weissman IL. Biology of hematopoietic stem cells and progenitors: implications for clinical application. Annu Rev Immunol. 2003;21:759–806. doi: 10.1146/annurev.immunol.21.120601.141007. [DOI] [PubMed] [Google Scholar]
  • 43.Fogg DK, Sibon C, Miled C, Jung S, Aucouturier P, Littman DR, Cumano A, Geissmann F. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science. 2006;311:83–87. doi: 10.1126/science.1117729. [DOI] [PubMed] [Google Scholar]
  • 44.Auffray C, Fogg DK, Narni-Mancinelli E, Senechal B, Trouillet C, Saederup N, Leemput J, Bigot K, Campisi L, Abitbol M, Molina T, Charo I, Hume DA, Cumano A, Lauvau G, Geissmann F. CX3CR1+ CD115+ CD135+ common macrophage/DC precursors and the role of CX3CR1 in their response to inflammation. J Exp Med. 2009;206:595–606. doi: 10.1084/jem.20081385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Onai N, Obata-Onai A, Schmid MA, Ohteki T, Jarrossay D, Manz MG. Identification of clonogenic common Flt3+ M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat Immunol. 2007;8:1207–1216. doi: 10.1038/ni1518. [DOI] [PubMed] [Google Scholar]
  • 46.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. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat Immunol. 2007;8:1217–1226. doi: 10.1038/ni1522. [DOI] [PubMed] [Google Scholar]
  • 47.Liu K, Victora GD, Schwickert TA, Guermonprez P, Meredith MM, Yao K, Chu FF, Randolph GJ, Rudensky AY, Nussenzweig M. In vivo analysis of dendritic cell development and homeostasis. Science. 2009;324:392–397. doi: 10.1126/science.1171243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med. 1973;137:1142–1162. doi: 10.1084/jem.137.5.1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. II. Functional properties in vitro. J Exp Med. 1974;139:380–397. doi: 10.1084/jem.139.2.380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Steinman RM, Lustig DS, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. 3. Functional properties in vivo. J Exp Med. 1974;139:1431–1445. doi: 10.1084/jem.139.6.1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Alvarez D, Vollmann EH, von Andrian UH. Mechanisms and consequences of dendritic cell migration. Immunity. 2008;29:325–342. doi: 10.1016/j.immuni.2008.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sunderkotter C, Nikolic T, Dillon MJ, Van Rooijen N, Stehling M, Drevets DA, Leenen PJ. Subpopulations of mouse blood monocytes differ in maturation stage and inflammatory response. J Immunol. 2004;172:4410–4417. doi: 10.4049/jimmunol.172.7.4410. [DOI] [PubMed] [Google Scholar]
  • 53.Varol C, Landsman L, Fogg DK, Greenshtein L, Gildor B, Margalit R, Kalchenko V, Geissmann F, Jung S. Monocytes give rise to mucosal, but not splenic, conventional dendritic cells. J Exp Med. 2007;204:171–180. doi: 10.1084/jem.20061011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Liu K, Waskow C, Liu X, Yao K, Hoh J, Nussenzweig M. Origin of dendritic cells in peripheral lymphoid organs of mice. Nat Immunol. 2007;8:578–583. doi: 10.1038/ni1462. [DOI] [PubMed] [Google Scholar]
  • 55.Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P, Figueiredo JL, Kohler RH, Chudnovskiy A, Waterman P, Aikawa E, Mempel TR, Libby P, Weissleder R, Pittet MJ. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009;325:612–616. doi: 10.1126/science.1175202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Serbina NV, Pamer EG. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat Immunol. 2006;7:311–317. doi: 10.1038/ni1309. [DOI] [PubMed] [Google Scholar]
  • 57.Tsou CL, Peters W, Si Y, Slaymaker S, Aslanian AM, Weisberg SP, Mack M, Charo IF. Critical roles for CCR2 and MCP-3 in monocyte mobilization from bone marrow and recruitment to inflammatory sites. J Clin Invest. 2007;117:902–909. doi: 10.1172/JCI29919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ishibashi M, Hiasa K, Zhao Q, Inoue S, Ohtani K, Kitamoto S, Tsuchihashi M, Sugaya T, Charo IF, Kura S, Tsuzuki T, Ishibashi T, Takeshita A, Egashira K. Critical role of monocyte chemoattractant protein-1 receptor CCR2 on monocytes in hypertension-induced vascular inflammation and remodeling. Circ Res. 2004;94:1203–1210. doi: 10.1161/01.RES.0000126924.23467.A3. [DOI] [PubMed] [Google Scholar]
  • 59.Drevets DA, Dillon MJ, Schawang JS, Van Rooijen N, Ehrchen J, Sunderkotter C, Leenen PJ. The Ly-6Chigh monocyte subpopulation transports Listeria monocytogenes into the brain during systemic infection of mice. J Immunol. 2004;172:4418–4424. doi: 10.4049/jimmunol.172.7.4418. [DOI] [PubMed] [Google Scholar]
  • 60.Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, Libby P, Weissleder R, Pittet MJ. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007;204:3037–3047. doi: 10.1084/jem.20070885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Swirski FK, Pittet MJ, Kircher MF, Aikawa E, Jaffer FA, Libby P, Weissleder R. Monocyte accumulation in mouse atherogenesis is progressive and proportional to extent of disease. Proc Natl Acad Sci USA. 2006;103:10340–10345. doi: 10.1073/pnas.0604260103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Swirski FK, Libby P, Aikawa E, Alcaide P, Luscinskas FW, Weissleder R, Pittet MJ. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest. 2007;117:195–205. doi: 10.1172/JCI29950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Combadiere C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, Merval R, Proudfoot A, Tedgui A, Mallat Z. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008;117:1649–1657. doi: 10.1161/CIRCULATIONAHA.107.745091. [DOI] [PubMed] [Google Scholar]
  • 64.Wu H, Gower RM, Wang H, Perrard XY, Ma R, Bullard DC, Burns AR, Paul A, Smith CW, Simon SI, Ballantyne CM. Functional role of CD11c+ monocytes in atherogenesis associated with hypercholesterolemia. Circulation. 2009;119:2708–2717. doi: 10.1161/CIRCULATIONAHA.108.823740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, Garin A, Liu J, Mack M, van Rooijen N, Lira SA, Habenicht AJ, Randolph GJ. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007;117:185–194. doi: 10.1172/JCI28549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Galkina E, Ley K. Immune and inflammatory mechanisms of atherosclerosis (*) Annu Rev Immunol. 2009;27:165–197. doi: 10.1146/annurev.immunol.021908.132620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Weber C, Belge KU, von Hundelshausen P, Draude G, Steppich B, Mack M, Frankenberger M, Weber KS, Ziegler-Heitbrock HW. Differential chemokine receptor expression and function in human monocyte subpopulations. J Leukoc Biol. 2000;67:699–704. doi: 10.1002/jlb.67.5.699. [DOI] [PubMed] [Google Scholar]
  • 68.Weber C, Zernecke A, Libby P. The multifaceted contributions of leukocyte subsets to atherosclerosis: lessons from mouse models. Nat Rev Immunol. 2008;8:802–815. doi: 10.1038/nri2415. [DOI] [PubMed] [Google Scholar]
  • 69.Jia T, Serbina NV, Brandl K, Zhong MX, Leiner IM, Charo IF, Pamer EG. Additive roles for MCP-1 and MCP-3 in CCR2-mediated recruitment of inflammatory monocytes during Listeria monocytogenes infection. J Immunol. 2008;180:6846–6853. doi: 10.4049/jimmunol.180.10.6846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Le Borgne M, Etcharts N, Goubier A, Lira SA, Sirard JC, van Rooijen N, Caux C, Ait-Yahia S, Vicari A, Kaiserlian D, Dubois B. Dendritic cells rapidly recruited into epithelial tissues via CCR6/CCL20 are responsible for CD8+ T cell crosspriming in vivo. Immunity. 2006;24:191–201. doi: 10.1016/j.immuni.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • 71.Gautier EL, Jakubzick C, Randolph GJ. Regulation of the migration and survival of monocyte subsets by chemokine receptors and its relevance to atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29:1412–1418. doi: 10.1161/ATVBAHA.108.180505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Saederup N, Chan L, Lira SA, Charo IF. Fractalkine deficiency markedly reduces macrophage accumulation and atherosclerotic lesion formation in CCR2−/− mice: evidence for independent chemokine functions in atherogenesis. Circulation. 2008;117:1642–1648. doi: 10.1161/CIRCULATIONAHA.107.743872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Combadiere C, Potteaux S, Gao JL, Esposito B, Casanova S, Lee EJ, Debre P, Tedgui A, Murphy PM, Mallat Z. Decreased atherosclerotic lesion formation in CX3CR1/apolipoprotein E double knockout mice. Circulation. 2003;107:1009–1016. doi: 10.1161/01.CIR.0000057548.68243.42. [DOI] [PubMed] [Google Scholar]
  • 74.Lesnik P, Haskell CA, Charo IF. Decreased atherosclerosis in CX3CR1−/− mice reveals a role for fractalkine in atherogenesis. J Clin Invest. 2003;111:333–340. doi: 10.1172/JCI15555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.An G, Wang H, Tang R, Yago T, McDaniel JM, McGee S, Huo Y, Xia L. P-selectin glycoprotein ligand-1 is highly expressed on Ly-6Chi monocytes and a major determinant for Ly-6Chi monocyte recruitment to sites of atherosclerosis in mice. Circulation. 2008;117:3227–3237. doi: 10.1161/CIRCULATIONAHA.108.771048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Libby P, Nahrendorf M, Pittet MJ, Swirski FK. Diversity of denizens of the atherosclerotic plaque: not all monocytes are created equal. Circulation. 2008;117:3168–3170. doi: 10.1161/CIRCULATIONAHA.108.783068. [DOI] [PubMed] [Google Scholar]
  • 77.Swirski FK, Weissleder R, Pittet MJ. Heterogeneous in vivo behavior of monocyte subsets in atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29:1424–1432. doi: 10.1161/ATVBAHA.108.180521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Woollard KJ, Geissmann F. Monocytes in atherosclerosis: subsets and functions. Nat Rev Cardiol. 2010;7:77–86. doi: 10.1038/nrcardio.2009.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Landsman L, Bar-On L, Zernecke A, Kim KW, Krauthgamer R, Shagdarsuren E, Lira SA, Weissman IL, Weber C, Jung S. CX3CR1 is required for monocyte homeostasis and atherogenesis by promoting cell survival. Blood. 2009;113:963–972. doi: 10.1182/blood-2008-07-170787. [DOI] [PubMed] [Google Scholar]
  • 80.Serbina NV, Jia T, Hohl TM, Pamer EG. Monocyte-mediated defense against microbial pathogens. Annu Rev Immunol. 2008;26:421–452. doi: 10.1146/annurev.immunol.26.021607.090326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol. 2007;82:244–252. doi: 10.1189/jlb.0307191. [DOI] [PubMed] [Google Scholar]
  • 82.Serbina NV, Salazar-Mather TP, Biron CA, Kuziel WA, Pamer EG. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity. 2003;19:59–70. doi: 10.1016/S1074-7613(03)00171-7. [DOI] [PubMed] [Google Scholar]
  • 83.Dunay IR, Damatta RA, Fux B, Presti R, Greco S, Colonna M, Sibley LD. Gr1(+) inflammatory monocytes are required for mucosal resistance to the pathogen Toxoplasma gondii. Immunity. 2008;29:306–317. doi: 10.1016/j.immuni.2008.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Barbalat R, Lau L, Locksley RM, Barton GM. Toll-like receptor 2 on inflammatory monocytes induces type I interferon in response to viral but not bacterial ligands. Nat Immunol. 2009;10:1200–1207. doi: 10.1038/ni.1792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Serbina NV, Kuziel W, Flavell R, Akira S, Rollins B, Pamer EG. Sequential MyD88-independent and -dependent activation of innate immune responses to intracellular bacterial infection. Immunity. 2003;19:891–901. doi: 10.1016/S1074-7613(03)00330-3. [DOI] [PubMed] [Google Scholar]
  • 86.Robben PM, LaRegina M, Kuziel WA, Sibley LD. Recruitment of Gr-1+ monocytes is essential for control of acute toxoplasmosis. J Exp Med. 2005;201:1761–1769. doi: 10.1084/jem.20050054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, Gherardi RK, Chazaud B. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med. 2007;204:1057–1069. doi: 10.1084/jem.20070075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, Sarnacki S, Cumano A, Lauvau G, Geissmann F. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science. 2007;317:666–670. doi: 10.1126/science.1142883. [DOI] [PubMed] [Google Scholar]
  • 89.Phillipson M, Heit B, Colarusso P, Liu L, Ballantyne CM, Kubes P. Intraluminal crawling of neutrophils to emigration sites: a molecularly distinct process from adhesion in the recruitment cascade. J Exp Med. 2006;203:2569–2575. doi: 10.1084/jem.20060925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ryschich E, Kerkadze V, Lizdenis P, Paskauskas S, Knaebel HP, Gross W, Gebhard MM, Buchler MW, Schmidt J. Active leukocyte crawling in microvessels assessed by digital time-lapse intravital microscopy. J Surg Res. 2006;135:291–296. doi: 10.1016/j.jss.2006.02.020. [DOI] [PubMed] [Google Scholar]
  • 91.Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. Development of monocytes, macrophages, and dendritic cells. Science. 2010;327:656–661. doi: 10.1126/science.1178331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Copin R, De Baetselier P, Carlier Y, Letesson JJ, Muraille E. MyD88-dependent activation of B220-CD11b+LY-6C+ dendritic cells during Brucella melitensis infection. J Immunol. 2007;178:5182–5191. doi: 10.4049/jimmunol.178.8.5182. [DOI] [PubMed] [Google Scholar]
  • 93.Narni-Mancinelli E, Campisi L, Bassand D, Cazareth J, Gounon P, Glaichenhaus N, Lauvau G. Memory CD8+ T cells mediate antibacterial immunity via CCL3 activation of TNF/ROI+ phagocytes. J Exp Med. 2007;204:2075–2087. doi: 10.1084/jem.20070204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Aldridge JRJ, Moseley CE, Boltz DA, Negovetich NJ, Reynolds C, Franks J, Brown SA, Doherty PC, Webster RG, Thomas PG. TNF/iNOS-producing dendritic cells are the necessary evil of lethal influenza virus infection. Proc Natl Acad Sci USA. 2009;106:5306–5311. doi: 10.1073/pnas.0900655106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Landsman L, Varol C, Jung S. Distinct differentiation potential of blood monocyte subsets in the lung. J Immunol. 2007;178:2000–2007. doi: 10.4049/jimmunol.178.4.2000. [DOI] [PubMed] [Google Scholar]
  • 96.Mantovani A, Garlanda C, Locati M. Macrophage diversity and polarization in atherosclerosis: a question of balance. Arterioscler Thromb Vasc Biol. 2009;29:1419–1423. doi: 10.1161/ATVBAHA.108.180497. [DOI] [PubMed] [Google Scholar]
  • 97.Martinez FO, Gordon S, Locati M, Mantovani A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol. 2006;177:7303–7311. doi: 10.4049/jimmunol.177.10.7303. [DOI] [PubMed] [Google Scholar]
  • 98.Lin SL, Castano AP, Nowlin BT, Lupher MLJ, Duffield JS. Bone marrow Ly6Chigh monocytes are selectively recruited to injured kidney and differentiate into functionally distinct populations. J Immunol. 2009;183:6733–6743. doi: 10.4049/jimmunol.0901473. [DOI] [PubMed] [Google Scholar]
  • 99.Dominguez PM, Ardavin C. Differentiation and function of mouse monocyte-derived dendritic cells in steady state and inflammation. Immunol Rev. 2010;234:90–104. doi: 10.1111/j.0105-2896.2009.00876.x. [DOI] [PubMed] [Google Scholar]
  • 100.Jakubzick C, Tacke F, Ginhoux F, Wagers AJ, van Rooijen N, Mack M, Merad M, Randolph GJ. Blood monocyte subsets differentially give rise to CD103+ and CD103− pulmonary dendritic cell populations. J Immunol. 2008;180:3019–3027. doi: 10.4049/jimmunol.180.5.3019. [DOI] [PubMed] [Google Scholar]
  • 101.Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt WD, Shakhar G, Jung S. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 2009;31:502–512. doi: 10.1016/j.immuni.2009.06.025. [DOI] [PubMed] [Google Scholar]
  • 102.Ginhoux F, Tacke F, Angeli V, Bogunovic M, Loubeau M, Dai XM, Stanley ER, Randolph GJ, Merad M. Langerhans cells arise from monocytes in vivo. Nat Immunol. 2006;7:265–273. doi: 10.1038/ni1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Chorro L, Sarde A, Li M, Woollard KJ, Chambon P, Malissen B, Kissenpfennig A, Barbaroux JB, Groves R, Geissmann F. Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network. J Exp Med. 2009;206:3089–3100. doi: 10.1084/jem.20091586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mildner A, Schmidt H, Nitsche M, Merkler D, Hanisch UK, Mack M, Heikenwalder M, Bruck W, Priller J, Prinz M. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci. 2007;10:1544–1553. doi: 10.1038/nn2015. [DOI] [PubMed] [Google Scholar]
  • 105.Bronte V, Apolloni E, Cabrelle A, Ronca R, Serafini P, Zamboni P, Restifo NP, Zanovello P. Identification of a CD11b(+)/Gr-1(+)/CD31(+) myeloid progenitor capable of activating or suppressing CD8(+) T cells. Blood. 2000;96:3838–3846. [PMC free article] [PubMed] [Google Scholar]
  • 106.Gallina G, Dolcetti L, Serafini P, De Santo C, Marigo I, Colombo MP, Basso G, Brombacher F, Borrello I, Zanovello P, Bicciato S, Bronte V. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest. 2006;116:2777–2790. doi: 10.1172/JCI28828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Movahedi K, Guilliams M, Van den Bossche J, Van den Bergh R, Gysemans C, Beschin A, De Baetselier P, Van Ginderachter JA. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood. 2008;111:4233–4244. doi: 10.1182/blood-2007-07-099226. [DOI] [PubMed] [Google Scholar]
  • 108.Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, Divino CM, Chen SH. Gr-1 + CD115 + immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006;66:1123–1131. doi: 10.1158/0008-5472.CAN-05-1299. [DOI] [PubMed] [Google Scholar]
  • 109.Ingersoll MA, Spanbroek R, Lottaz C, Gautier EL, Frankenberger M, Hoffmann R, Lang R, Haniffa M, Collin M, Tacke F, Habenicht AJ, Ziegler-Heitbrock L, Randolph GJ. Comparison of gene expression profiles between human and mouse monocyte subsets. Blood. 2010;115:e10–e19. doi: 10.1182/blood-2009-07-235028. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Life Sciences: CMLS are provided here courtesy of Springer

RESOURCES