Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Oct 9.
Published in final edited form as: Front Biosci (Landmark Ed). 2009 Jan 1;14:2631–2646. doi: 10.2741/3401

THE CONTRIBUTION OF TYRO3 FAMILY RECEPTOR TYROSINE KINASES TO THE HETEROGENEITY OF APOPTOTIC CELL UPTAKE BY MONONUCLEAR PHAGOCYTES

Jeffrey L Curtis 1, Jill C Todt 1, Bin Hu 1, John J Osterholzer 1, Christine M Freeman 1
PMCID: PMC4191832  NIHMSID: NIHMS589919  PMID: 19273223

Abstract

Mononuclear phagocytes comprise a mobile, broadly dispersed and highly adaptable system that lies at the very epicenter of host defense against pathogens and the interplay of the innate and adaptive arms of immunity. Understanding the molecular mechanisms that control the response of mononuclear phagocytes to apoptotic cells and the anti-inflammatory consequences of that response is an important goal with implications for multiple areas of biomedical sciences. This review details current understanding of the heterogeneity of apoptotic cell uptake by different members of the mononuclear phagocyte family in humans and mice. It also recounts the unique role of the Tyro3 family of receptor tyrosine kinases, best characterized for Mertk, in the signal transduction leading both to apoptotic cell ingestion and the anti-inflammatory effects that result.

Keywords: Apoptosis; Dendritic cells; Innate Immunity; Macrophages; Phagocytosis; Receptor Tyrosine Kinase; Review, tutorial

2. INTRODUCTION - THE IMPORTANCE OF MONONUCLEAR PHAGOCYTE UPTAKE OF APOPTOTIC CELLS

Clearance of apoptotic cells is important during embryonic development and in the maintenance of immunological self-tolerance 1, but becomes crucial during the resolution of infection 2-5. During successfully controlled infections, large numbers of leukocytes die by apoptosis 6-8, and their prompt, efficient clearance by tissue macrophages safely disposes of the proteolytic enzymes that they contain. Tissue repair is hastened by mediators released by macrophages during apoptotic cell ingestion 9-13, which include transforming growth factor-β (TGF-β), platelet activating factor (PAF), prostaglandin E2 (PGE2) and interleukin (IL-)10 14-17. In a contact-dependent fashion that is incompletely understood, macrophage recognition of apoptotic cells also suppresses production of inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), granulocyte-macrophage colony stimulating factor, IL-1β, IL-12 and IL-18 18-23. However, these same anti-inflammatory mediators released during apoptotic cell ingestion by macrophages have properties that can compromise host defenses against pathogens and promote fibrosis 24, 25. By contrast, if apoptotic cells are not promptly ingested, they progress to a late apoptotic stage that is similar to necrosis, lose membrane integrity, and disintegrate 26. Necrotic cells and late apoptotic cells drive maturation of immature dendritic cells and hence T cell activation 27-29. Inflammation can be perpetuated when fragments of late apoptotic cells, notably those containing High Mobility Group Box-1 protein, bind to Toll-like receptor-4 (TLR4), better known as the receptor for purified lipopolysaccharide (LPS), thereby activating NF-κB 9, 30. Uningested late apoptotic cells also release nucleosomes, which are formed in vivo exclusively by chromatin digestion during apoptosis. Nucleosomes are a major immunogen for autoantibodies in systemic lupus erythematosus 31-35 and are implicated in atherogenesis 36-38. Thus, efficient apoptotic cell clearance contributes centrally to protective adaptive immunity, appropriate resolution of innate inflammation, and the prevention of autoimmunity.

Macrophage-independent mechanisms for apoptotic cell clearance exist 39-42. Epithelial cells in the thymus and in the involuting female breast avidly ingest cells undergoing apoptosis in their respective organs 43-46. Lung epithelial cells and fibroblasts ingest apoptotic cells in vitro (albeit less efficiently than do macrophages) 47-49. Although the overall importance of apoptotic cell uptake by these cell types in vivo is unproven, it is plausible that they contribute significantly in the absence of overt inflammation. However, during steady state in most organs, and certainly during inflammation, mononuclear phagocytes are believed to clear most apoptotic cells. These considerations imply that understanding the molecular mechanisms controlling the response of mononuclear phagocytes to apoptotic cells and its consequences is an important goal with implications for multiple areas of biomedical sciences.

This review details the current understanding of heterogeneity of apoptotic cell uptake by different members of the mononuclear phagocyte family, and the central role in that process of one family of receptor tyrosine kinases (RTKs), the Tyro3 family. Because portions of this subject have been reviewed previously 50-53, emphasis will be on recent developments and their potential implications for human health and disease, and on unresolved questions. We also discuss why Tyro3 RTKs are so crucial for the signal transduction leading both to ingestion and to the anti-inflammatory effects that result.

3. HETEROGENEITY OF APOPTOTIC CELL UPTAKE BY MONONUCLEAR PHAGOCYTES

Mononuclear phagocytes are a heterogeneous group of hematopoietic cells that inhabit every organ 54-56. In the most general of terms, the mononuclear phagocyte system consists of circulating monocytes, tissue macrophages and dendritic cells. Except for the plasmacytoid dendritic cell subset, all appear to derive from a common precursor 57, 58.

3.1. Monocytes

Monocytes begin their journey by emigrating from the bone marrow into the blood in a fashion that depends on CC chemokine receptor 2 (CCR2) under both normal and inflammatory conditions 59, 60. CCR2 binds several CC chemokines, include CCL2 (formerly known as MCP- 1), a chemokine that specifically induces monocyte chemotaxis. Monocytes circulate briefly before migrating to tissues where they differentiate into macrophages or dendritic cells 61. Some controversy persists regarding the potential of individual monocytes to differentiate into these two differentiated mononuclear phagocyte types; this controversy relates to the issue of monocytes subsets.

There is a long history of attempting to subdivide monocytes into subsets that possess different capacities for migration and subsequent differentiation. Human monocytes comprise two principal subsets, a majority “classical” CD14high CD16- population and a “non-classical” or “proinflammatory” CD14low CD16+ population 55, 62, 63; a third, very minor double-positive subset has also been identified 64. The CD16- subset, but not the CD16+ subset of human monocytes can differentiate in vitro into osteoclasts 65. In a model of reverse-transendothelial migration 66, the monocytes that developed characteristics of dendritic cells were predominately CD16+ 67. However, both subsets of human monocyte could differentiate into dendritic cells in culture when stimulated by cytokines 68. The degree to which each of these monocyte subsets gives rise to other specialized mononuclear phagocyte cell types in non-lymphoid tissue is incompletely characterized.

Evidence from a transgenic model system suggests that circulating monocytes in mice also comprise two distinct phenotypes. These phenotypes can be distinguished based on their expression of two surface molecules, the CX3C chemokine receptor CX3CR1, which is the unique receptor for the chemokine CX3CL1 (fractalkine), and the Gr-1 epitope found on the Ly-6C/G family of glycosylphosphatidylinositol-linked molecules on myeloid cells. These two monocyte phenotypes are, respectively, a short-lived CX3CR1low CCR2+ Gr-1high subset that can be recruited to sites of inflammation, and a longer-lived CX3CR1high CCR2 Gr-1low “resident subset” that constitutively migrates to a variety of tissues in a CX3CR1-dependent fashion 69. The CX3CR1high monocyte subset was recently shown to crawl along vascular endothelia in a fashion that depends on LFA-1 (CD11a/CD18), permitting them very rapid entry into sites of inflammation 70. Importantly, CX3CR1 expression also distinguishes the two main subsets of monocytes in the rat 71, and in humans, where the CD14low CD16+ population expresses higher levels 69. Thus, an emerging paradigm holds that mammalian monocytes can be divided into separate lineages distinguishable by their level of expression of CX3CR1.

Sunderkotter et al. modified this concept somewhat 72, in a study that analyzed murine monocytes using the ER-MP20 monoclonal antibody, which specifically recognizes only Ly-6C, a macrophage-specific protein, and not Ly-6G, which is also expressed by other myeloid cells. Because the previous study by Geissman et al. 69 had defined Gr-1 expression using the monoclonal antibody RB6-8C5, which recognizes an epitope common to Ly-6G and Ly-6C 73, direct comparison between the two studies is difficult. Sunderkotter et al. showed that Ly-6Chigh monocytes were recent bone marrow emigrants, which could be recruited to sites of inflammation, and that they gave rise to the more mature Ly-6Clow monocytes, which had lost this ability 72. The Ly-6Clow subset also preferentially expressed CD11c and the anti-adhesive molecule CD43. They further showed a continuum of Ly-6C expression between Ly-6Chigh and Ly-6Clow subsets, with a transient Ly-6Cintermediate population. These data imply a developmental rather than parallel lineage relationship between the two monocyte phenotypes. In other studies, both the Gr-1high (Ly-6C/Ghigh) and the Gr-1low murine monocyte subsets were originally reported to be able to differentiate into dendritic cells in vivo, but their ability to differentiate into macrophages in vivo was unknown. A later study focusing on the pulmonary mononuclear phagocyte system confirmed that both monocyte subsets could differentiate into lung dendritic cells, but that only Gr-1low monocytes had the potential to differentiate immediately into lung macrophages 74.

The ability to ingest apoptotic cells is gained as monocytes differentiate within tissues. Monocytes freshly isolated from the human peripheral blood have very limited capacity to ingest apoptotic cells 75, 76. Incubation in serum or in TGF-β induces some ability for human monocytes to ingest apoptotic cells, generally with a marked dependence on CD36 and vitronectin 77. Because differentiation of monocytes into the wide variety of macrophages and dendritic cells found in individual organs appears to be under complex and likely unique regulation, these seminal data will need re-examination as the conditions for organ-specific differentiation are defined. Like most other aspects of monocyte biology in the mouse, the capacity of murine peripheral blood monocytes to ingest apoptotic cells is largely unknown, due to the difficulty in isolating sufficient numbers of this cell type at high purity. To investigate whether monocytes newly recruited to a site of inflammation contribute to apoptotic cell clearance, we used in situ vital-staining of resident tissue macrophages in a well-characterized model of CD4 T cell– dependent lung inflammation 78. Compared to resident alveolar macrophages, monocytes recruited to the lung ingested apoptotic cells poorly, whereas they avidly ingested immunoglobulin-opsonized particles. In this antigen-driven, non-infectious model system, overall numbers of resident alveolar macrophages did not change significantly over the course of the response and the majority of recruited monocytes did not become long-term residents. By contrast, in LPS-induced lung inflammation, Maus et al. found high alveolar macrophage turnover and persistent replacement by immigrating monocytes 79. These differences in results likely relate to the degree to which the inflammatory process induces apoptosis of resident tissue macrophages, a characteristic feature of some infections 80-88

3.2. Resident tissue macrophages

The phenotype of resident macrophages in different organs obviously differs very markedly, spanning the range from hepatic Kuppfer cells to microglial cells of the central nervous system. Resident tissue macrophages derive ultimately from hematopoietic precursors, but there is evidence that in the absence of inflammation in some organs, their numbers are sustained primarily by local proliferation 89-92. In the mouse, the resident peritoneal macrophage is often taken to be the “generic” macrophage, due to the ease of its isolation. Peritoneal macrophages bind apoptotic cells readily by means of multiple receptors and ingest apoptotic cells fairly avidly, making them an ideal reference cell type. In peritoneal macrophages and likely in most mononuclear phagocyte cell types, a crucial signal for ingestion is stereospecific recognition of externalized phosphatidylserine (PS) on the apoptotic cell surface 93, 94 via one or more receptors that are blocked, in mice, by monoclonal antibody 217 95. The biochemical nature of stereospecific PS recognition remains elusive 96.

Other than the peritoneal macrophage, the most thoroughly studied resident macrophage is probably the alveolar macrophage, the principal resident phagocyte of the lungs. Resident alveolar macrophages show markedly reduced apoptotic cell uptake in vitro relative to resident peritoneal macrophages (in the mouse) 97, 98 or relative to alveolar macrophages harvested from rabbits three days after the induction of immune-complex-induced injury 75. The reduced phagocytosis in alveolar macrophages was attributable in part to decreased adhesion of the apoptotic cells 98, as well as to markedly decreased expression of protein kinase C (PKC) βII, the sole isoform of PKC required for apoptotic cell uptake 99. Although the pulmonary collectin surfactant protein A increased apoptotic cell ingestion by alveolar macrophages 100, 101, the effect was minor and did not increase ingestion by alveolar macrophages to the levels seen using peritoneal macrophages 98. Human resident alveolar macrophages also show low capacity to ingest apoptotic cells 102-105. Interestingly, resident human alveolar macrophages also have very decreased expression of PKC βII, relative to monocytes 106.

Some data on this issue also exist for microglia, the resident mononuclear phagocyte of the central nervous system. Microglial cells from surgically resected normal human or rat brains ingested autologous apoptotic cells, which they bound using CD36 and recognize via externalized PS 3, 107, 108. Little is currently known about apoptotic cell uptake by resident macrophages in other tissues, especially for humans.

Mature tissue macrophages can also assume a variety of activation states that have been characterized as classical activation in response to interferon-gamma, and alternative activation (reviewed in 109, 110). The capacities of macrophages in these different activation states to ingest apoptotic cells has received only scant attention 111. Polarized macrophages (induced in vitro from human peripheral blood CD14+ monocytes supplemented with Granulocyte-Colony Stimulating Factor) showed increased ability to bind and ingest apoptotic cells, and unique ability to ingest cells in the early stages of apoptosis 112. Additional studies examining this issue via polarization of resident tissue macrophages from various organs would contribute to the understanding of specific disease states.

3.3. Dendritic cells

Dendritic cells are a heterogeneous population of antigen-presenting cells with complex ontogeny, but distinct dendritic cells subsets can be identified based upon anatomic location, migratory pathways, and specific immunological function 113. In the resting state, two major dendritic cells lineages can be distinguished: conventional dendritic cells and plasmacytoid dendritic cells. Conventional dendritic cells were previously called myeloid dendritic cells, a term that is falling from usage since it was found that conventional dendritic cells can develop from both myeloid and lymphoid precursors 114.

Conventional dendritic cells display a characteristic stellate morphology and participate in classical dendritic cell functions including uptake, processing, and presentation of antigens to naïve and memory T cells. Shortman and Naik divide conventional dendritic cells into “lymphoid-tissue-resident” and “migratory” populations 113. Lymphoid-tissue-resident conventional dendritic cells include three subsets within the spleen or lymph nodes (CD4+/CD8−, CD4−/CD8+, and CD4-/CD8-) and up to four additional conventional dendritic cells populations in the thymus. Migratory dendritic cells include epidermal Langerhans cells 115 and the interstitial dendritic cells of non-lymphoid organs, including the dermis, liver, and the intestinal, respiratory and reproductive tracts. In response to inflammatory stimuli, migratory conventional dendritic cells travel in a CCR7-dependent manner to draining lymph nodes, where they present antigen to naïve T cells.

Whilst migratory conventional dendritic cells are exiting peripheral tissues during inflammation, in the mouse they are replaced by inflammatory dendritic cells that are derived from CX3CR1lo CCR2+ Gr-1+ inflammatory monocytes (as described above) and possibly from CD11c+ dendritic cell precursors in the blood. Gene-targeting has confirmed the essential role of the chemokine receptor CCR2 in the recruitment of inflammatory dendritic cells to the skin, spleen and lungs 116-118. Inflammatory dendritic cells resemble resting migratory conventional dendritic cells in phenotype, although it is uncertain whether they also migrate to regional nodes to sustain antigen presentation, or are predominately active within the inflamed tissues. The latter possibility is supported by experiments in mice showing dendritic cell-dependent local production of TNF-α and inducible nitric oxide synthase in response to Listeria monocytogenes 59 and by promotion of tissue-specific TH1 polarization by IL-12 production in response to Cryptococcus neoformans (Osterholzer et al., manuscript submitted).

The efficiency and functional consequence of the uptake of apoptotic cells by conventional dendritic cells depends not only on the dendritic cells subset, but also on the micro-anatomic location and context in which the apoptotic cell is encountered (i.e., resting vs. inflammatory conditions) 119. Uptake of apoptotic cells within lymphoid structures (including lymph nodes and the thymus) and by immature migratory conventional dendritic cells without simultaneous stimulation via pathogen recognition receptors is believed to contribute crucially to maintenance of T-cell tolerance 120-126. This concept has been challenged recently by the finding that apoptotic cells derived from polyclonally-activated peripheral blood mononuclear cells (PBMC), but not those from resting PBMC, could induce maturation of human monocyte-derived dendritic cells 127. In contrast, uptake of a late apoptotic cell (or necrotic cell) induces conventional dendritic cells to mature 122, 128, to migrate to local lymph nodes (for migratory dendritic cells), and to stimulate T cell activation. Although this process may enhance host defense against microbial antigens, it also risks presentation of self antigens to T cells in a stimulatory context and the subsequent generation of autoimmunity.

Plasmacytoid dendritic cells comprise an independent dendritic cell lineage 129-131. Formed primarily in the bone marrow, these dendritic cells take up residence in both lymphoid and non-lymphoid organs. They produce large amounts of type-1 interferon in response to a variety of microbial stimuli and are, therefore, particularly important to anti-viral immunity (although conventional dendritic cells also may serve a role). Whether plasmacytoid dendritic cells participate in the clearance of apoptotic cells has not been established. Dalgaard et al. compared the ability of myeloid dendritic cells and plasmacytoid dendritic cells to ingest apoptotic leukemic cells and concluded that plasmacytoid dendritic cells do not contribute to apoptotic cell uptake 132. However, Hoeffel et al. demonstrated that plasmacytoid dendritic cells acquisition of antigens from cells undergoing viral-induced apoptosis promoted cross presentation to CD8+ T cells 133. Whether these were late apoptotic cells (and thus more stimulatory than the leukemic apoptotic cells studied by Dalgaard et al.) cannot be determined.

4. THE TYRO3 FAMILY OF RTKs

The mammalian Tyro3 family consists of three closely related receptor tyrosine kinases (RTKs) that are involved in apoptotic cell uptake and immunomodulation. The three individual family members have acquired a variety of names, reflecting their initial independent identification by multiple laboratories. Table 1 lists the names used in this review (Tyro3, Axl and Mertk), together with common synonyms and identifiers for the human and murine genes. Variable amounts of all three RTKs are expressed by normal and malignant cells of neural, lymphoid vascular and reproductive origin 134. Tyro3 family members were originally identified for their transforming ability when over-expressed 135-139, and as such, can be considered protooncogenes. Indeed, this concept has been supported by over-expression in both murine and human systems, and several human cancers over-express Mertk (summarized in 140). All three RTKs are involved in megakaryocytopoiesis and platelet aggregation 140-142. Because blockade of Tyro3 RTKs inhibits thrombosis without enhancing bleeding, this activity is under intense scrutiny from the pharmaceutical industry. Axl is also implicated in natural killer cell differentiation and vascular remodeling 143. It is likely that the complete range of properties of this interesting molecular family remains undefined.

Table 1.

Names, synonyms & gene ID of Tyro3 RTKs

Name Synonyms GeneID1 OMIM2
Tyro3 Sky, Rse, Bit, DTK, Tif, Etk-2 7301 (h) 600341
22174 (m)
Axl Ark, Ufo, Tyro7, AI323647 558 (h) 109135
26362 (m)
Mertk Tyro12, c-mer, Eyk, Nyk, RP23-30M20.1 10461 (h) 604705
17289 (m)
1

(h) = human gene, (m) = murine gene, Entrez Gene Home (http://www.ncbi.nlm.nih.gov)

2

OMIM, Online Mendelian Inheritance in Man

4.1. Structure & function of Tyro3 RTKs

Tyro3 family RTKs are defined by a unique shared sequence (in single letter amino acid code, KWIAIES) within their kinase domain. Tyro3 is prominently expressed in the developing brain, as well as in adult kidney, testis and ovary, and by both human pulmonary artery endothelial cells and osteoclasts. Axl appears to be the most widely distributed member of the family, due to its expression in cell lines of epithelial, mesenchymal and hematopoietic origins as well as non-transformed cells 135. Mertk was originally named c-mer due to its expression in monocytes, epithelial cells, and reproductive tissue 144.

Tyro3 RTKs are single-pass type 1 transmembrane proteins with characteristic extra- and intracellular modular structures. All three appear to be heavily glycosylated; Mertk has 13 potential N-linked glycosylation sites. The extracellular region of each RTK includes two amino terminal immunoglobulin motifs followed by two more membrane proximal fibronectin type-III motifs. Immediately after molecular cloning of these RTKs, it was suggested that these motifs might participate in cell-cell or cell-matrix interactions 137. Axl was shown to mediate homophilic binding leading to cell aggregation when over-expressed in S2 cells 145, but to date, there is no direct in vivo experimental evidence for this attractive possibility. The alternative possibility, that these motifs contribute to binding of Tyro3 RTKs in cis to other macrophage cell surface receptors, is supported by evidence for a pairing of Axl (but not Tyro3 or Mertk) with the IL-15Rα chain 146. This interaction required the extracellular portion of Axl and was present in the absence of ligand stimulation. Treatment with IL-15 induced tyrosine phosphorylation of both Axl and IL-15Rα , and activation of the phosphatidylinositol-3 kinase (PI-3K)-Akt pathway, indicative of the functional nature of this heterologous receptor pairing. These data illustrate the capacity of Tyro3 RTKs to serve as signaling molecules in association with other receptors, a possibility to which we will return.

All three Tyro3 family RTKs are receptors for the serum protein growth arrest-specific gene 6 (Gas6), whereas only Tyro3 binds protein S. Both Gas6 and protein S are vitamin K-dependent soluble factors that can bind externalized PS 147-149, the hallmark of an apoptotic cell. Binding of these soluble factors provides a means by which Tyro3 family RTKs could contribute to apoptotic cell recognition. However, Biacore assay has shown that the association of Mertk with Gas6 is of lower avidity than that of Axl and Tyro3 147. This finding suggests that Mertk may depend to a greater degree than other family members on association with other apoptotic cell-binding receptors to initiate apoptotic cell ingestion. One way in which this could occur is via heterodimerization of Mertk with other Tyro3 RTKs, as has recently been suggested 150. The spatial relationships of Axl binding to Gas6 was recently clarified by analysis of crystal structure at 3.3Å resolution of a minimal human Gas6-Axl complex. Results revealed an assembly of 2:2 stoichiometry, in which the two immunoglobulin-like domains of Axl were cross-linked by the first laminin G-like domain of Gas6, without direct Axl-Axl or Gas6-Gas6 contacts. This study also found major and minor Gas6 binding sites on Axl, and showed that both binding sites were required for productive transmembrane signaling. Interestingly, only the minor Gas6 binding site is highly conserved in Tyro3 and Mertk 151.

The complexity of ligand-binding by Tyro3 RTKs is increased by the finding that soluble Axl can be generated by ADAM10-dependent cleavage, and is found in high concentration in mouse serum in association with Gas6 152. Although no such soluble forms of murine Tyro3 or Mertk were detected in that study, a soluble form of human Mertk was predicted based on an alternatively spliced RNA transcript 137. Soluble Mertk was recently found to be produced by multiple human macrophage cell types, and to inhibit both macrophage clearance of apoptotic cells and platelet aggregation 140.

4.2. Difference in Tyro3 RTK expression among mononuclear phagocyte subsets

Tyro3 family RTKs were first suspected to contribute to apoptotic cell clearance based on observations in two separate systems: gene-targeted mice lacking expression of all three RTKs, which showed blindness and massive apoptotic cell accumulation in the spleen 134; and positional cloning of a gene involved in retinitis pigmentosa in rats 153, 154, which identified a spontaneous mutation in Mertk. Blindness in the rats proved to result from defective ingestion of photoreceptor outer segments by retinal pigment epithelial cells, a process closely akin to apoptotic cell uptake 155. The central role of these RTKs in apoptotic cell uptake was thereafter rapidly and conclusively confirmed using the gene-targeted mice. Triple knockout mice lacking all three Tyro3 RTKs developed a severe lymphoproliferative disorder characterized by auto-immunity and widespread thrombosis associated with anti-phospholipid antibodies 156. Mice lacking Mertk expression showed accumulation of uningested apoptotic thymocytes after injection of dexamethasone, and reduced clearance of labeled apoptotic cells from the spleen and thioglycollate-treated peritoneal cavity, following intravenous and intraperitoneal injection, respectively 157.

Of the three Tyro3 RTKs, monocytes appear to express mRNA only for Mertk, the family member most restricted to the mononuclear phagocyte lineage 137, 138. Using flow cytometry, we found that mononuclear phagocytes freshly-recruited to the lungs express only very low amounts of Mertk 78. This observation, in agreement with unpublished data from Behrens et al. 158, is not necessarily at odds with the original finding that Mertk could be detected on circulating human monocytes, as that earlier data relied on RT-PCR 137.

Tissue resident macrophages may differ in expression of individual Tyro3 RTKs {e.g., resident murine alveolar macrophages appear to express all three receptors, whereas resident peritoneal macrophages from unstimulated mice express only MerTK (unpublished observation)}, although this subject has not been defined for many organs in either mice or humans. In the primary tissue macrophages for which it has been examined, dependence on Mertk for apoptotic cell uptake is significant 78, 98, 157, whereas the role for Tyro3 and Axl appears more minor 150. To our knowledge, expression of Tyro3 RTKs and their contribution to apoptotic cell uptake have not been examined in relation to polarized states of macrophage activation in either species.

The role of Tyro3 RTKs in apoptotic cell uptake by murine dendritic cells has been investigated by two groups. Behrens et al. found that splenic dendritic cells of both conventional (CD11c+, CD11b+) and CD8αα+ subsets expressed Mertk, whereas the absence of Mertk+ cells in B cell zones of the spleen argued against expression by follicular dendritic cells 158. In that study, bone marrow-derived dendritic cells from Mertk-deficient mice showed no impairment in uptake of apoptotic cells. Similarly, Mertk was shown by Seitz et al. to be unnecessary for apoptotic cell uptake by bone marrow-derived dendritic cells and splenic dendritic cells. By contrast, they found reduced apoptotic cell ingestion by bone marrow-derived dendritic cells, splenic dendritic cells, and to a lesser degree thioglycollate-elicited peritoneal exudate macrophages from mice lacking either Axl, Tyro3 or both RTKs 150. There are currently no data on this subject in humans or in resident dendritic cells from mucosal surfaces in mice.

The differential capacity of tissue macrophages versus dendritic cells to ingest apoptotic cells 50 has important consequences for immunoregulation. For example, because immature dendritic cells but not macrophages can induce a specific cytotoxic T cell response after ingesting virally-infected apoptotic cells 120, development of specific anti-viral immunity depends on the cell type of the mononuclear phagocyte that ingests the infected apoptotic cells.

5. TYRO3 RTKS AS SIGNAL-TRANSDUCING ELEMENTS DURING APOPTOTIC CELL UPTAKE

5.1. Basics of signal-transduction during apoptotic cell uptake

Apoptotic cell recognition by mononuclear phagocytes activates an intracellular signaling cascade leading to efficient apoptotic cell engulfment. This engulfment process was originally believed to be similar to phagocytosis triggered by Fc receptors (FcR), and the two processes do share some characteristics, such as dependence on PI-3K and phospholipase C γ (PLC γ) 159, 160. However, both biochemical and morphological differences between uptake of the two types of particles are now appreciated. PKC βII is uniquely required for apoptotic cell ingestion 99, which differs from the PKC isoform requirements for macrophage phagocytosis of other particles 161, 162. Apoptotic cell ingestion does not require the cytoplasmic tyrosine kinase Syk 163, which is essential for FcγR-mediated ingestion 164, 165. FcR-mediated uptake involves “membrane zippering” 166, whereas apoptotic cell uptake has been show to involve macro-pinocytosis, leading to formation of spacious phagosomes 112, 167, 168 (although this point has been contested 169-171). The unique features of the apoptotic cell engulfment process have earned it a new term, “efferocytosis” (from effero — to carry to the grave) 172.

Because absence or inhibition of Tyro3 RTKs has no effect on FcR-mediated phagocytosis, to understand their role in apoptotic cell clearance it is helpful to consider the minimal signaling pathways essential for that process, which are strictly conserved between worms (such as Caenorhabdiis elegans) and mammals (Fig. 1). Genetic analysis in C. elegans 173 has identified two crucial epistatic pathways. One pathway consists of the cell surface receptor ced-1 (likely mammalian homolog: lipoprotein receptor-related protein, also known as CD91), the intracellular adapter ced-6 (GULP), and the ATP-binding cassette transporter ced-7 (mammalian homolog recently shown to be ABCA7 174). The other pathway consists of the adaptor protein ced-2 (mammalian homolog CrkII), the adaptor protein ced-5 (Dock180), the Rho family GTP-binding protein ced-10 (Rac1), and the Rho family GTP-binding protein ced-12 (ELMO) 175-177. The ced-1, -6, -7 pathway also acts genetically upstream of ced-10 in worms 178, but the biochemical point of connection between the two pathways is not yet established.

Figure 1. Schematic diagram of the epistatic pathways of essential genes for apoptotic cell clearance in worms, with presumed mammalian homologs.

Figure 1

The engulfing cell (for C. elegans) or macrophage is shown in outline, and the apoptotic cell (AC) is in gray. The names of C. elegans genes are shown in black letters and those of the corresponding mammalian genes are in blue letters. The proteins of the CED-1/CED-6/ CED-7 pathway are shown in red. Note that CED-7 must be present on both the apoptotic cell & the engulfing cell. The proteins of the CED-2/CED-5/CED-10/CED-12 pathway are in green. Arrows indicate actin cytoskeleton-driven membrane reorganization leading to engulfment. Adapted from 175

The ced-2, -5, -10, -12 pathway (CrkII-Dock180-Rac-ELMO in mammals), which is entirely intracellular, is required for actin cytoskeleton restructuring during phagocytosis. This pathway is regulated by RhoG, which increases apoptotic cell engulfment, and RhoA, which decreases it 179. In keeping with this finding, RhoG and its exchange factor TRIO were found to interact with ELMO, promoting Rac1 activation and cytoskeletal reorganization 180. However, exactly how this intracellular pathway interacts with the many surface receptors involved in apoptotic cell engulfment remains incompletely defined. Until recently, the only known link was with the αvβ5 integrin, which must interact with the p130Cas-CrkII-Dock180 complex for apoptotic cell internalization 181. It was also known that inhibitors of protein tyrosine kinases blocked apoptotic cell uptake 159, 181 by both dendritic cells, which use αvβ5 integrin, and macrophages, which use αvβ3 integrin 121.

5.2. Features of Tyro3 RTKs that facilitate signal transduction

The properties of Tyro3 family members (reviewed in 53) imply that they could interact with many other macrophage molecules to transduce signals during apoptotic cell ingestion. Besides their tyrosine kinase activity, such properties include their ability to serve as cell surface receptors for soluble PS-binding proteins, their multiple extracellular motifs reminiscent of adhesion molecules, and their intracellular multi-ligand domain. Such a signaling role is supported by several types of data.

First, prompt and specific tyrosine phosphorylation of Mertk itself is induced by exposure to apoptotic cells (for macrophages) or outer rod segments (for retinal pigment epithelial cells) 160, 182 . This finding matches the suspected mechanism of activation of most RTKs by ligand-induced oligomerization, typically dimerization. Oligomerization yields an active kinase via auto-transphosphorylation of tyrosines in the kinase activation loop or juxtamembrane region 183. Indeed, a chimeric molecule containing the transmembrane and intracellular domains of Mertk showed ligand-inducible tyrosine autophosphorylation 184. RTK tyrosine phosphorylation is also needed to form binding sites within the intracellular multi-ligand domain for SH2-domain containing proteins.

Second, the intracellular multi-ligand domain of Tyro3 family RTKS have been shown to bind many signaling intermediaries known or suspected to be involved for apoptotic cell engulfment. For example, interaction of Tyro3 with its ligand protein S recruits Src 185, and the p85 subunit of phosphatidylinositol-3-OH kinase (PI-3K) 186, which is required for apoptotic cell ingestion 159. Axl can bind PI-3K, PLC γ, Grb2, Src, and others, presumably via their SH2 domains. Axl also binds C1-TEN (C1 domain-containing phosphatase and TENsin homologue), a protein without other currently identified partners 187, 188.

Third, dimerization of Mertk during apoptotic cell recognition leads to activation of downstream mediators. One example is the tyrosine phosphorylation of PLC γ 160, a key signaling event that leads to formation of inositol 1,4,5 trisphosphate and diacylglycerol, which are essential for cytoskeletal rearrangements. Similarly, ligand-dependent activation of Mertk on human monocytes induces phosphorylation and release from MerTK of the guanine exchange factor Vav1, and subsequent activation of the RhoA family members Rac1, Cdc42 and RhoA 189. This constitutive, SH2-dependent, but phosphotyrosine-independent interaction between Mertk and Vav1 is unusual. The cytoplasmic domain of Mertk has also been shown to be able to tyrosine phosphorylate PI-3K, Shc and Grb2 190. It is likely that ligand-induced oligomerization occurs during apoptotic cell recognition by other Tyro3 family receptors, but there is also potential for ligand-independent dimerization 151, 191.

Mertk so far appears unique in its ability to alter the cytoskeleton on apoptotic cell binding through small G protein regulators of cytoskeletal dynamics such as Rac1, Cdc42 and RhoA 184. The Birge laboratory has shown that Mertk activation (by Gas6 or via a constitutively active Mertk construct) induces Src-mediated phosphorylation of focal adhesion kinase (FAK), which in turn recruits FAK to the αvβ5 integrin. The ultimate result is Rac1 activation via the 130CAS-CrkII-Dock180 complex 192. This finding is important because it indicates involvement of Mertk in the classical apoptotic cell signaling pathway (Fig. 1). Conversely, during ingestion of photoreceptor outer segment fragments by retinal pigment epithelial cells, phagocytosis and signaling involving MerTK were abolished by absence either of αvβ5 integrin or of the apoptotic cell-binding soluble factor milk fat globule-EGF-factor 8 (MFG-E8), which bind αvβ5 integrin but not Mertk 193. Taken together, these findings imply that neither Mertk nor αvβ5 integrin can be considered simply to be downstream of the other. Instead, the two molecules appear to cooperate to mediate apoptotic cell recognition. It is likely that Mertk interacts with additional transmembrane molecules, e.g., we have recently found evidence that scavenger receptor A I/II (CD204) is essential for optimal activation of MerTK and subsequent signaling required for apoptotic cell ingestion by murine macrophages (Todt et al., submitted).

5.3. What's so special about Tyro3 RTKs?

A wide variety of macrophage surface receptors from unrelated molecular families have been implicated in apoptotic cell ingestion 10, 101, 194-203. Why so many receptors are involved in this process is unresolved. One viewpoint holds that redundancy is essential to safeguard such a crucial process. This explanation is supported by macrophage recognition of ICAM-3 on apoptotic neutrophils but not lymphocytes 204, 205. Alternatively, Gregory and Devitt have suggested that this redundancy increases the flexibility of the system, by allowing a more specific response to the context in which the apoptosis occurs 206. Multiple low-affinity receptor-ligand interactions may jointly serve as a high avidity complex, similar to the immunological synapse formed between T cells and antigen-presenting cells.

A corollary of this viewpoint, that receptors from different molecular families could serve different and fundamentally unique functions, is particularly relevant to the Tyro3 RTKs. Among the bewildering array of receptors implicated in apoptotic cell uptake, the evidence for involvement of many comes only from antibody blocking experiments that did not distinguish between a role in apoptotic cell adhesion and a signaling role essential for ingestion 167. Importantly, adhesion of apoptotic cells to a variety of murine tissue macrophages and macrophage cell lines is not inhibited by function-blocking monoclonal antibody against Mertk 98; whether this is the case for Axl and Tyro3 in other mononuclear phagocyte subsets is unknown. Normal tethering of apoptotic cells is seen in macrophages and dendritic cells from Mertk deficient mice and in macrophages from transgenic mice lacking MFG-E8, both of which show highly defective in vivo apoptotic cell clearance 157, 207. Further support for an essential signaling role for Tyro3 RTKs comes from the observation that an overt autoimmune phenotype results from deletion of only a subset of the many genes implicated in apoptotic cell clearance. Intriguingly, Tyro3 RTKs are among them 134, 208, along with MFG-E8 209, serum amyloid P-component, C1q, C4 and IgM 176.

Tyro3 RTKs, especially Mertk, also appear to contribute to the immunosuppressive effect of apoptotic cell engagement. Mice lacking Mertk surface expression displayed hypersensitivity to LPS in vivo that could be largely reversed by anti-TNF-α. Relative to peritoneal exudate macrophages from wild-type mice, peritoneal exudate macrophages from these mutant mice over-produced TNF-α and showed increased and sustained binding of NF-kB to the TNF-α promoter 210. Splenic dendritic cells from triple-mutant mice lacking expression of all three Tyro3 RTKs exhibited markedly increased expression of major histocompatibility complex class II molecules and CD86 after in vitro LPS stimulation 156. Mertk is also required for apoptotic cell-induced inhibition of NF-kB activation by LPS in murine bone marrow-derived dendritic cells 211. Collectively, these findings could imply that signals emanating from Tyro3 RTKS (or possibly just Mertk) intersect signals arising from the TLR4/CD14 complex. This exciting possibility clearly merits additional study. Interestingly, Ebola and Marburg viruses use Tyro3 RTKs to gain cell entry 212, which has been suggested to contribute to the dysregulation of the immune system and vascular endothelium seen during infection with these highly pathogenic filoviruses.

A significant increase in the understanding of how Tyro3 RTKs contribute to the immunomodulatory effect of apoptotic cells came from the recent finding that activation of Axl by Gas6 or apoptotic cells induces Twist proteins 1 and 2 213, which are basic helix-loop-helix transcriptional repressors that inhibit production of TNF-α and other NF-κB-dependent inflammatory cytokines 214. In a separate study, the typical anti-inflammatory response in macrophages ingesting apoptotic cells was not seen when the target was itself a macrophage induced to apoptosis by free-cholesterol loading (but not by ultraviolet irradiation or exposure to oxidized low density lipoproteins) of elicited murine peritoneal macrophages 215. In this case, there was induction of TNF-α and IL-6 that was reduced in the absence of Mertk, but not of CD91, another receptor implicated in apoptotic cell uptake. Inflammatory cytokine release required cell-cell contact and the ERK 1/2 signaling pathway, but neither apoptotic cell ingestion nor the TLR adapter MyD88. These findings implicate Mertk as a potential therapeutic target in late in atherosclerotic plaques.

6. CONCLUSIONS

Given the complex ontogeny of tissue macrophages and dendritic cells, it is simplistic to make broad generalizations about apoptotic cell uptake of these two cell types without reference to their source and its state of inflammation. The phenotype of tissue macrophages and dendritic cells residing in different organs has probably been selected during evolution to provide an optimal balance between the potentially immunosuppressive effects of apoptotic cell ingestion (relative to that organ's exposure to pathogens) and the potentially damaging effects of delayed apoptotic cell clearance. Differences in expression by mononuclear phagocytes of the Tyro3 RTKs, and possibly differences in the association of Tyro3 RTKs with other receptors, are undoubtedly a significant component of this evolutionary process. By contrast, monocytes recruited into these tissues appear to start out largely without ability to ingest apoptotic cells, and hence appear to be highly malleable by the environment to which they are recruited.

The essential role of Tyro3 family RTKs in apoptotic cell uptake by mononuclear phagocytes likely results because they can perform two crucial functions simultaneously: (a) providing assembly points for a broad range of signaling intermediaries; (b) activating multiple molecules crucial for actin cytoskeleton restructuring. Tyro3 RTKs probably also contribute to form the phagocyte:apoptotic cell synapse because they are cell surface receptors for soluble PS-binding proteins, and possibly because their extracellular motifs can interact with adhesion molecules on the apoptotic cell, or with other receptors on the phagocyte surface. Because Tyro3 RTKs appear to be unique among receptors for apoptotic cells in displaying this range of contributions, we hazard the prediction that they will prove to be the central “trigger” through which the apoptotic cell ingestion process is initiated.

Until the factors controlling differentiation of specific macrophage and dendritic cell subsets from monocytes are better defined, results from analyses of mononuclear phagocyte cell lines and in vitro-manipulated monocytes must be validated against the actual behavior of macrophages and dendritic cells in different tissues and in various disease states. This validation process, far from being “descriptive”, is a crucial step in the translation of basic science into approaches that can be tested in clinical research. A robust molecular understanding of apoptotic cell clearance by specific subsets of mononuclear phagocytes holds great promise for novel therapeutic approaches to most chronic diseases.

ACKNOWLEDGEMENTS

This work was supported by Merit Review funds, a Career Development Award and a Research Enhancement Award Program (REAP) from the Biomedical Laboratory Research & Development Service, Department of Veterans Affairs; and by RO1 HL056309, R01 HL082480 and T32 HL07749 from NIH. We thank Drs. Gary D. Luker, Bethany B. Moore, Robert Paine, III, and Galen B. Toews for helpful discussion, and Dr. Luker for critiquing the manuscript.

Abbreviations

FAK

focal adhesion kinase

FcR

Fc receptor(s)

Gas6

growth arrest-specific gene 6

IL-

interleukin

LPS

lipopolysaccharide

MFG-E8

milk fat globule-EGF-factor 8, also known as lactadherin

PAF

platelet activating factor

PBMC

peripheral blood mononuclear cells

PGE2

prostaglandin E2

PKC

protein kinase C

PLC

phospholipase C

PS

phosphatidylserine

RTK

receptor tyrosine kinase

TGF-β

transforming growth factor-beta

TLR4

Toll-like receptor-4

TNF-α

tumor necrosis factor-alpha

REFERENCES

  • 1.Henson PM, Hume DA. Apoptotic cell removal in development and tissue homeostasis. Trends Immunol. 2006;27:244–50. doi: 10.1016/j.it.2006.03.005. [DOI] [PubMed] [Google Scholar]
  • 2.Haslett C. Granulocyte apoptosis and its role in the resolution and control of lung inflammation. Am J Respir Crit Care Med. 1999;160:S5–S11. doi: 10.1164/ajrccm.160.supplement_1.4. [DOI] [PubMed] [Google Scholar]
  • 3.Chan A, Seguin R, Magnus T, Papadimitriou C, Toyka KV, Antel JP, Gold R. Phagocytosis of apoptotic inflammatory cells by microglia and its therapeutic implications: termination of CNS autoimmune inflammation and modulation by interferon-beta. Glia. 2003;43:231–42. doi: 10.1002/glia.10258. [DOI] [PubMed] [Google Scholar]
  • 4.Huynh ML, Malcolm KC, Kotaru C, Tilstra JA, Westcott JY, Fadok VA, Wenzel SE. Defective apoptotic cell phagocytosis attenuates prostaglandin E2 and 15-hydroxyeicosatetraenoic acid in severe asthma alveolar macrophages. Am J Respir Crit Care Med. 2005;172:972–9. doi: 10.1164/rccm.200501-035OC. [DOI] [PubMed] [Google Scholar]
  • 5.Erwig L-P, Henson P. Clearance of apoptotic cells by phagocytes. Cell Death Differ. 2008;15:243–250. doi: 10.1038/sj.cdd.4402184. [DOI] [PubMed] [Google Scholar]
  • 6.Akbar AN, Savill J, Gombert W, Bofill M, Borthwick NJ, Whitelaw F, Grundy J, Janossy G, Salmon M. The specific recognition by macrophages of CD8+,CD45RO+ T cells undergoing apoptosis: A mechanism for T cell clearance during resolution of viral infections. J Exp Med. 1994;180:1943–1947. doi: 10.1084/jem.180.5.1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Razvi ES, Jiang Z, Woda BA, Welsh RM. Lymphocyte apoptosis during the silencing of the immune response to acute viral infections in normal, lpr, and Bcl-2-transgenic mice. Am J Pathol. 1995;147:79–91. [PMC free article] [PubMed] [Google Scholar]
  • 8.Chen M, Wang YH, Wang Y, Huang L, Sandoval H, Liu YJ, Wang J. Dendritic cell apoptosis in the maintenance of immune tolerance. Science. 2006;311:1160–4. doi: 10.1126/science.1122545. [DOI] [PubMed] [Google Scholar]
  • 9.Li M, Carpio DF, Zheng Y, Bruzzo P, Singh V, Ouaaz F, Medzhitov RM, Beg AA. An essential role of the NF-kappa B/Toll-like receptor pathway in induction of inflammatory and tissue-repair gene expression by necrotic cells. J Immunol. 2001;166:7128–35. doi: 10.4049/jimmunol.166.12.7128. [DOI] [PubMed] [Google Scholar]
  • 10.Savill J, Dransfield I, Gregory C, Haslett C. A blast from the past: clearance of apoptotic cells regulates immune responses. Nat Rev Immunol. 2002;2:965–75. doi: 10.1038/nri957. [DOI] [PubMed] [Google Scholar]
  • 11.Liu CY, Liu YH, Lin SM, Yu CT, Wang CH, Lin HC, Lin CH, Kuo HP. Apoptotic neutrophils undergoing secondary necrosis induce human lung epithelial cell detachment. J Biomed Sci. 2003;10:746–56. doi: 10.1007/BF02256327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Curtis JL. Cell-mediated adaptive immune defense of the lungs. Proc Am Thorac Soc. 2005;2:412–6. doi: 10.1513/pats.200507-070JS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rose DM, Sydlaske AD, Agha-Babakhani A, Johnson K, Terkeltaub R. Transglutaminase 2 limits murine peritoneal acute gout-like inflammation by regulating macrophage clearance of apoptotic neutrophils. Arthritis Rheum. 2006;54:3363–71. doi: 10.1002/art.22137. [DOI] [PubMed] [Google Scholar]
  • 14.Freire-de-Lima CG, Nacimento DO, Soares MBP, Bozza PT, Castro-Faria-Neto HC, de Mello FG, DosReis GA, Lopes MF. Uptake of apoptotic cells drives the growth of a pathogenic trypanosome in macrophages. Nature. 2000;403:199–203. doi: 10.1038/35003208. [DOI] [PubMed] [Google Scholar]
  • 15.Huynh ML, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. J Clin Invest. 2002;109:41–50. doi: 10.1172/JCI11638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Byrne A, Reen DJ. Lipopolysaccharide induces rapid production of IL-10 by monocytes in the presence of apoptotic neutrophils. J Immunol. 2002;168:1968–1977. doi: 10.4049/jimmunol.168.4.1968. [DOI] [PubMed] [Google Scholar]
  • 17.Freire-de-Lima CG, Xiao YQ, Gardai SJ, Bratton DL, Schiemann WP, Henson PM. Apoptotic cells, through Transforming Growth Factor-beta, coordinately induce anti-inflammatory and suppress pro-inflammatory eicosanoid and NO synthesis in murine macrophages. J Biol Chem. 2006;281:38376–84. doi: 10.1074/jbc.M605146200. [DOI] [PubMed] [Google Scholar]
  • 18.Lucas M, Stuart LM, Savill J, Lacy-Hulbert A. Apoptotic cells and innate immune stimuli combine to regulate macrophage cytokine secretion. J Immunol. 2003;171:2610–5. doi: 10.4049/jimmunol.171.5.2610. [DOI] [PubMed] [Google Scholar]
  • 19.Kim S, Elkon KB, Ma X. Transcriptional suppression of interleukin-12 gene expression following phagocytosis of apoptotic cells. Immunity. 2004;21:643–53. doi: 10.1016/j.immuni.2004.09.009. [DOI] [PubMed] [Google Scholar]
  • 20.Chung EY, Kim SJ, Ma XJ. Regulation of cytokine production during phagocytosis of apoptotic cells. Cell Res. 2006;16:154–61. doi: 10.1038/sj.cr.7310021. [DOI] [PubMed] [Google Scholar]
  • 21.Lucas M, Stuart LM, Zhang A, Hodivala-Dilke K, Febbraio M, Silverstein R, Savill J, Lacy-Hulbert A. Requirements for apoptotic cell contact in regulation of macrophage responses. J Immunol. 2006;177:4047–54. doi: 10.4049/jimmunol.177.6.4047. [DOI] [PubMed] [Google Scholar]
  • 22.Cvetanovic M, Mitchell JE, Patel V, Avner BS, Su Y, van der Saag PT, Witte PL, Fiore S, Levine JS, Ucker DS. Specific recognition of apoptotic cells reveals a ubiquitous and unconventional innate immunity. J Biol Chem. 2006;281:20055–20067. doi: 10.1074/jbc.M603920200. [DOI] [PubMed] [Google Scholar]
  • 23.Patel VA, Longacre A, Hsiao K, Fan H, Meng F, Mitchell JE, Rauch J, Ucker DS, Levine JS. Apoptotic cells, at all stages of the death process, trigger characteristic signaling events that are divergent from and dominant over those triggered by necrotic cells: Implications for the delayed clearance model of autoimmunity. J Biol Chem. 2006;281:4663–70. doi: 10.1074/jbc.M508342200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Tridandapani S, Wardrop R, Baran CP, Wang Y, Opalek JM, Caligiuri MA, Marsh CB. TGF-beta 1 suppresses myeloid Fc gamma receptor function by regulating the expression and function of the common gamma-subunit. J Immunol. 2003;170:4572–7. doi: 10.4049/jimmunol.170.9.4572. [DOI] [PubMed] [Google Scholar]
  • 25.Aronoff DM, Canetti C, Peters-Golden M. Prostaglandin E2 inhibits alveolar macrophage phagocytosis through an E-prostanoid 2 receptor-mediated increase in intracellular cyclic AMP. J Immunol. 2004;173:559–65. doi: 10.4049/jimmunol.173.1.559. [DOI] [PubMed] [Google Scholar]
  • 26.Wyllie AH, Kerr JF, Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol. 1980;68:251–306. doi: 10.1016/s0074-7696(08)62312-8. [DOI] [PubMed] [Google Scholar]
  • 27.Sauter B, Albert ML, Francisco L, Larsson M, Somersan S, Bhardwaj N. Consequences of cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J Exp Med. 2000;191:423–434. doi: 10.1084/jem.191.3.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Barker RN, Erwig L, Pearce WP, Devine A, Rees AJ. Differential effects of necrotic or apoptotic cell uptake on antigen presentation by macrophages. Pathobiology. 1999;67:302–5. doi: 10.1159/000028085. [DOI] [PubMed] [Google Scholar]
  • 29.Rovere-Querini P, Capobianco A, Scaffidi P, Valentinis B, Catalanotti F, Giazzon M, Dumitriu IE, Muller S, Iannacone M, Traversari C, Bianchi ME, Manfredi AA. HMGB1 is an endogenous immune adjuvant released by necrotic cells. EMBO Rep. 2004;5:825–30. doi: 10.1038/sj.embor.7400205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, Mignot G, Maiuri MC, Ullrich E, Saulnier P, Yang H, Amigorena S, Ryffel B, Barrat FJ, Saftig P, Levi F, Lidereau R, Nogues C, Mira JP, Chompret A, Joulin V, Clavel-Chapelon F, Bourhis J, Andre F, Delaloge S, Tursz T, Kroemer G, Zitvogel L. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–1059. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
  • 31.Bell DA, Morrison B, VandenBygaart P. Immunogenic DNA-related factors. Nucleosomes spontaneously released from normal murine lymphoid cells stimulate proliferation and immunoglobulin synthesis of normal mouse lymphocytes. J Clin Invest. 1990;85:1487–96. doi: 10.1172/JCI114595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Morrison SL. In vitro antibodies: strategies for production and application. Annu Rev Immunol. 1992;10:239–65. doi: 10.1146/annurev.iy.10.040192.001323. [DOI] [PubMed] [Google Scholar]
  • 33.Mohan C, Adams S, Stanik V, Datta SK. Nucleosome: a major immunogen for pathogenic autoantibody-inducing T cells of lupus. J Exp Med. 1993;177:1367–81. doi: 10.1084/jem.177.5.1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Burlingame RW, Boey ML, Starkebaum G, Rubin RL. The central role of chromatin in autoimmune responses to histones and DNA in systemic lupus erythematosus. J Clin Invest. 1994;94:184–92. doi: 10.1172/JCI117305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mevorach D, Zhou JL, Song X, Elkon KB. Systemic exposure to irradiated apoptotic cells induces autoantibody production. J Exp Med. 1998;188:387–92. doi: 10.1084/jem.188.2.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gordon S. Do macrophage innate immune receptors enhance atherogenesis? Dev Cell. 2003;5:666–8. doi: 10.1016/s1534-5807(03)00329-0. [DOI] [PubMed] [Google Scholar]
  • 37.Aprahamian T, Rifkin I, Bonegio R, Hugel B, Freyssinet JM, Sato K, Castellot JJ, Jr., Walsh K. Impaired clearance of apoptotic cells promotes synergy between atherogenesis and autoimmune disease. J Exp Med. 2004;199:1121–31. doi: 10.1084/jem.20031557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Schrijvers DM, De Meyer GR, Kockx MM, Herman AG, Martinet W. Phagocytosis of apoptotic cells by macrophages is impaired in atherosclerosis. Arterioscler Thromb Vasc Biol. 2005;25:1256–61. doi: 10.1161/01.ATV.0000166517.18801.a7. [DOI] [PubMed] [Google Scholar]
  • 39.Fadok VA. Clearance: the last and often forgotten stage of apoptosis. J Mammary Gland Biol Neoplasia. 1999;4:203–11. doi: 10.1023/a:1011384009787. [DOI] [PubMed] [Google Scholar]
  • 40.Hall MO, Prieto AL, Obin MS, Abrams TA, Burgess BL, Heeb MJ, Agnew BJ. Outer segment phagocytosis by cultured retinal pigment epithelial cells requires Gas6. Exp Eye Res. 2001;73:509–20. doi: 10.1006/exer.2001.1062. [DOI] [PubMed] [Google Scholar]
  • 41.Wiegand UK, Corbach S, Prescott AR, Savill J, Spruce BA. The trigger to cell death determines the efficiency with which dying cells are cleared by neighbours. Cell Death Differ. 2001;8:734–46. doi: 10.1038/sj.cdd.4400867. [DOI] [PubMed] [Google Scholar]
  • 42.Nakanishi Y, Shiratsuchi A. Phagocytic removal of apoptotic spermatogenic cells by Sertoli cells: mechanisms and consequences. Biol Pharm Bull. 2004;27:13–6. doi: 10.1248/bpb.27.13. [DOI] [PubMed] [Google Scholar]
  • 43.Hiramine C, Nakagawa T, Miyauchi A, Hojo K. Thymic nurse cells as the site of thymocyte apoptosis and apoptotic cell clearance in the thymus of cyclophosphamide-treated mice. Lab Invest. 1996;75:185–201. [PubMed] [Google Scholar]
  • 44.Small M, Kraal G. In vitro evidence for participation of DEC-205 expressed by thymic cortical epithelial cells in clearance of apoptotic thymocytes. Int Immunol. 2003;15:197–203. doi: 10.1093/intimm/dxg024. [DOI] [PubMed] [Google Scholar]
  • 45.Cao WM, Murao K, Imachi H, Hiramine C, Abe H, Yu X, Dobashi H, Wong NC, Takahara J, Ishida T. Phosphatidylserine receptor cooperates with high-density lipoprotein receptor in recognition of apoptotic cells by thymic nurse cells. J Mol Endocrinol. 2004;32:497–505. doi: 10.1677/jme.0.0320497. [DOI] [PubMed] [Google Scholar]
  • 46.Monks J, Rosner D, Jon Geske F, Lehman L, Hanson L, Neville MC, Fadok VA. Epithelial cells as phagocytes: apoptotic epithelial cells are engulfed by mammary alveolar epithelial cells and repress inflammatory mediator release. Cell Death Differ. 2005;12:107–14. doi: 10.1038/sj.cdd.4401517. [DOI] [PubMed] [Google Scholar]
  • 47.Walsh GM, Sexton DW, Blaylock MG, Convery CM. Resting and cytokine-stimulated human small airway epithelial cells recognize and engulf apoptotic eosinophils. Blood. 1999;94:2827–35. [PubMed] [Google Scholar]
  • 48.Sexton DW, Blaylock MG, Walsh GM. Human alveolar epithelial cells engulf apoptotic eosinophils by means of integrin- and phosphatidylserine receptor-dependent mechanisms: a process upregulated by dexamethasone. J Allergy Clin Immunol. 2001;108:962–9. doi: 10.1067/mai.2001.119414. [DOI] [PubMed] [Google Scholar]
  • 49.Hall SE, Savill JS, Henson PM, Haslett C. Apoptotic neutrophils are phagocytosed by fibroblasts with participation of the fibroblast vitronectin receptor and involvement of a mannose/fucose-specific lectin. J Immunol. 1994;153:3218–27. [PubMed] [Google Scholar]
  • 50.Xu W, Roos A, Daha MR, van Kooten C. Dendritic cell and macrophage subsets in the handling of dying cells. Immunobiology. 2006;211:567–75. doi: 10.1016/j.imbio.2006.05.023. [DOI] [PubMed] [Google Scholar]
  • 51.Tietzel I, Mosser DM. The modulation of macrophage activation by tyrosine phosphorylation. Front Biosci. 2002;7:d1494–502. doi: 10.2741/tietzel. [DOI] [PubMed] [Google Scholar]
  • 52.Lemke G, Lu Q. Macrophage regulation by Tyro 3 family receptors. Curr Opin Immunol. 2003;15:31–6. doi: 10.1016/s0952-7915(02)00016-x. [DOI] [PubMed] [Google Scholar]
  • 53.Hafizi S, Dahlback B. Signalling and functional diversity within the Axl subfamily of receptor tyrosine kinases. Cytokine Growth Factor Rev. 2006;17:295–304. doi: 10.1016/j.cytogfr.2006.04.004. [DOI] [PubMed] [Google Scholar]
  • 54.Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, Gordon S. Macrophage receptors and immune recognition. Annu Rev Immunol. 2005;23:901–44. doi: 10.1146/annurev.immunol.23.021704.115816. [DOI] [PubMed] [Google Scholar]
  • 55.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5:953–64. doi: 10.1038/nri1733. [DOI] [PubMed] [Google Scholar]
  • 56.Hume DA. The mononuclear phagocyte system. Curr Opin Immunol. 2006;18:49–53. doi: 10.1016/j.coi.2005.11.008. [DOI] [PubMed] [Google Scholar]
  • 57.del Hoyo GM, Martin P, Vargas HH, Ruiz S, Arias CF, Ardavin C. Characterization of a common precursor population for dendritic cells. Nature. 2002;415:1043–7. doi: 10.1038/4151043a. [DOI] [PubMed] [Google Scholar]
  • 58.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;311:83–7. doi: 10.1126/science.1117729. [DOI] [PubMed] [Google Scholar]
  • 59.Serbina NV, Pamer EG. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat Immunol. 2006;7:311–7. doi: 10.1038/ni1309. [DOI] [PubMed] [Google Scholar]
  • 60.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;117:902–9. doi: 10.1172/JCI29919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lehtonen A, Ahlfors H, Veckman V, Miettinen M, Lahesmaa R, Julkunen I. Gene expression profiling during differentiation of human monocytes to macrophages or dendritic cells. J Leukoc Biol. 2007;82:710–20. doi: 10.1189/jlb.0307194. [DOI] [PubMed] [Google Scholar]
  • 62.Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol. 2007;82:244–52. doi: 10.1189/jlb.0307191. [DOI] [PubMed] [Google Scholar]
  • 63.Ziegler-Heitbrock L. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leukoc Biol. 2007;81:584–92. doi: 10.1189/jlb.0806510. [DOI] [PubMed] [Google Scholar]
  • 64.Passlick B, Flieger D, Ziegler-Heitbrock HW. Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood. 1989;74:2527–34. [PubMed] [Google Scholar]
  • 65.Komano Y, Nanki T, Hayashida K, Taniguchi K, Miyasaka N. Identification of a human peripheral blood monocyte subset that differentiates into osteoclasts. Arthritis Res Ther. 2006;8:R152. doi: 10.1186/ar2046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Randolph GJ, Beaulieu S, Lebecque S, Steinman RM, Muller WA. Differentiation of monocytes into dendritic cells in a model of transendothelial trafficking. Science. 1998;282:480–3. doi: 10.1126/science.282.5388.480. [DOI] [PubMed] [Google Scholar]
  • 67.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;196:517–27. doi: 10.1084/jem.20011608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.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;13:1571–81. doi: 10.1093/intimm/13.12.1571. [DOI] [PubMed] [Google Scholar]
  • 69.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]
  • 70.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;317:666–70. doi: 10.1126/science.1142883. [DOI] [PubMed] [Google Scholar]
  • 71.Yrlid U, Jenkins CD, MacPherson GG. Relationships between distinct blood monocyte subsets and migrating intestinal lymph dendritic cells in vivo under steady-state conditions. J Immunol. 2006;176:4155–62. doi: 10.4049/jimmunol.176.7.4155. [DOI] [PubMed] [Google Scholar]
  • 72.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;172:4410–7. doi: 10.4049/jimmunol.172.7.4410. [DOI] [PubMed] [Google Scholar]
  • 73.Fleming TJ, Fleming ML, Malek TR. Selective expression of Ly-6G on myeloid lineage cells in mouse bone morrow. RB6-8C5 mAb to granulocyte-differentiation antigen (Gr-1) detects members of the Ly-6 family. J Immunol. 1983;151:2399–2408. [PubMed] [Google Scholar]
  • 74.Landsman L, Varol C, Jung S. Distinct differentiation potential of blood monocyte subsets in the lung. J Immunol. 2007;178:2000–7. doi: 10.4049/jimmunol.178.4.2000. [DOI] [PubMed] [Google Scholar]
  • 75.Newman SL, Henson JE, Henson PM. Phagocytosis of senescent neutrophils by human monocyte-derived macrophages and rabbit inflammatory macrophages. J Exp Med. 1982;156:430–42. doi: 10.1084/jem.156.2.430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Savill J, Dransfield I, Hogg N, Haslett C. Vitronectin receptor-mediated phagocytosis of cells undergoing apoptosis. Nature. 1990;343:170–173. doi: 10.1038/343170a0. [DOI] [PubMed] [Google Scholar]
  • 77.Ren Y, Savill J. Proinflammatory cytokines potentiate thrombospondin-mediated phagocytosis of neutrophils undergoing apoptosis. J Immunol. 1995;154:2366–74. [PubMed] [Google Scholar]
  • 78.Jennings JH, Linderman DJ, Hu B, Sonstein J, Curtis JL. Monocytes recruited to the lungs of mice during immune inflammation ingest apoptotic cells poorly. Am J Respir Cell Mol Biol. 2005;32:108–117. doi: 10.1165/rcmb.2004-0108OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Maus UA, Janzen S, Wall G, Srivastava M, Blackwell TS, Christman JW, Seeger W, Welte T, Lohmeyer J. Resident alveolar macrophages are replaced by recruited monocytes in response to endotoxin-induced lung inflammation. Am J Respir Cell Mol Biol. 2006;35:227–235. doi: 10.1165/rcmb.2005-0241OC. [DOI] [PubMed] [Google Scholar]
  • 80.Tateda K, Deng JC, Moore TA, Newstead MW, Paine R, 3rd, Kobayashi N, Yamaguchi K, Standiford TJ. Hyperoxia mediates acute lung injury and increased lethality in murine Legionella pneumonia: the role of apoptosis. J Immunol. 2003;170:4209–16. doi: 10.4049/jimmunol.170.8.4209. [DOI] [PubMed] [Google Scholar]
  • 81.Knapp S, Leemans JC, Florquin S, Branger J, Maris NA, Pater J, van Rooijen N, van der Poll T. Alveolar macrophages have a protective antiinflammatory role during murine pneumococcal pneumonia. Am J Respir Crit Care Med. 2003;167:171–9. doi: 10.1164/rccm.200207-698OC. [DOI] [PubMed] [Google Scholar]
  • 82.Dockrell DH, Marriott HM, Prince LR, Ridger VC, Ince PG, Hellewell PG, Whyte MK. Alveolar macrophage apoptosis contributes to pneumococcal clearance in a resolving model of pulmonary infection. J Immunol. 2003;171:5380–8. doi: 10.4049/jimmunol.171.10.5380. [DOI] [PubMed] [Google Scholar]
  • 83.Ruckdeschel K, Pfaffinger G, Haase R, Sing A, Weighardt H, Hacker G, Holzmann B, Heesemann J. Signaling of apoptosis through TLRs critically involves toll/IL-1 receptor domain-containing adapter inducing IFN-beta, but not MyD88, in bacteria-infected murine macrophages. J Immunol. 2004;173:3320–8. doi: 10.4049/jimmunol.173.5.3320. [DOI] [PubMed] [Google Scholar]
  • 84.DeLeo FR. Modulation of phagocyte apoptosis by bacterial pathogens. Apoptosis. 2004;9:399–413. doi: 10.1023/B:APPT.0000031448.64969.fa. [DOI] [PubMed] [Google Scholar]
  • 85.Zhang Y, Bliska JB. Role of macrophage apoptosis in the pathogenesis of Yersinia. Curr Top Microbiol Immunol. 2005;289:151–73. doi: 10.1007/3-540-27320-4_7. [DOI] [PubMed] [Google Scholar]
  • 86.Marriott HM, Hellewell PG, Cross SS, Ince PG, Whyte MK, Dockrell DH. Decreased alveolar macrophage apoptosis is associated with increased pulmonary inflammation in a murine model of pneumococcal pneumonia. J Immunol. 2006;177:6480–8. doi: 10.4049/jimmunol.177.9.6480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Dockrell DH, Whyte MK. Regulation of phagocyte lifespan in the lung during bacterial infection. J Leukoc Biol. 2006;79:904–8. doi: 10.1189/jlb.1005555. [DOI] [PubMed] [Google Scholar]
  • 88.Finlay BB, McFadden G. Anti-immunology: evasion of the host immune system by bacterial and viral pathogens. Cell. 2006;124:767–82. doi: 10.1016/j.cell.2006.01.034. [DOI] [PubMed] [Google Scholar]
  • 89.Chan WY, Kohsaka S, Rezaie P. The origin and cell lineage of microglia: new concepts. Brain Res Rev. 2007;53:344–54. doi: 10.1016/j.brainresrev.2006.11.002. [DOI] [PubMed] [Google Scholar]
  • 90.Sawyer RT. The ontogeny of pulmonary alveolar macrophages in parabiotic mice. J Leukoc Biol. 1986;40:347–54. doi: 10.1002/jlb.40.4.347. [DOI] [PubMed] [Google Scholar]
  • 91.Shellito J, Esparza C, Armstrong C. Maintenance of the normal rat alveolar macrophage cell population: the roles of monocyte influx and alveolar macrophage proliferation in situ. Am Rev Respir Dis. 1987;135:78–82. doi: 10.1164/arrd.1987.135.1.78. [DOI] [PubMed] [Google Scholar]
  • 92.Evans MJ, Sherman MP, Campbell LA, Shami SG. Proliferation of pulmonary alveolar macrophages during postnatal development of rabbit lungs. Am Rev Respir Dis. 1987;136:384–7. doi: 10.1164/ajrccm/136.2.384. [DOI] [PubMed] [Google Scholar]
  • 93.Fadok VA, Voelker DR, Campbell PA, Cohen JJ, Bratton DL, Henson PM. Exposure of phophatidylserine on the surface of apoptotic lymphocytes triggers specific recognition and removal by macrophages. J Immunol. 1992;148:2207–2216. [PubMed] [Google Scholar]
  • 94.Fadok VA, Bratton DL, Frasch SC, Warner ML, Henson PM. The role of phosphatidylserine in recognition of apoptotic cells by phagocytes. Cell Death Differ. 1998;5:551–62. doi: 10.1038/sj.cdd.4400404. [DOI] [PubMed] [Google Scholar]
  • 95.Fadok VA, Bratton DL, Rose DM, Pearson A, Ezekewitz RA, Henson PM. A receptor for phosphatidylserine-specific clearance of apoptotic cells. Nature. 2000;405:85–90. doi: 10.1038/35011084. [DOI] [PubMed] [Google Scholar]
  • 96.Schlegel RA, Williamson P. P.S. to PS (phosphatidylserine)--pertinent proteins in apoptotic cell clearance. Sci STKE. 2007:pe57. doi: 10.1126/stke.4082007pe57. 2007. [DOI] [PubMed] [Google Scholar]
  • 97.Hu B, Sonstein J, Christensen PJ, Punturieri A, Curtis JL. Deficient in vitro and in vivo phagocytosis of apoptotic T cells by resident murine alveolar macrophages. J Immunol. 2000;165:2124–2133. doi: 10.4049/jimmunol.165.4.2124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Hu B, Jennings JH, Sonstein J, Floros J, Todt JC, Polak T, Curtis JL. Resident murine alveolar and peritoneal macrophages differ in adhesion of apoptotic thymocytes. Am J Respir Cell Mol Biol. 2004;30:687–693. doi: 10.1165/rcmb.2003-0255OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Todt JC, Hu B, Punturieri A, Sonstein J, Polak T, Curtis JL. Activation of Protein Kinase C beta II by the stereo-specific phosphatidylserine receptor is required for phagocytosis of apoptotic thymocytes by resident murine tissue macrophages. J Biol Chem. 2002;277:35906–35914. doi: 10.1074/jbc.M202967200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Schagat TL, Wofford JA, Wright JR. Surfactant protein A enhances alveolar macrophage phagocytosis of apoptotic neutrophils. J Immunol. 2001;166:2727–2733. doi: 10.4049/jimmunol.166.4.2727. [DOI] [PubMed] [Google Scholar]
  • 101.Vandivier RW, Ogden CA, Fadok VA, Hoffmann PR, Brown KK, Botto M, Walport MJ, Fisher JH, Henson PM, Greene KE. Role of surfactant proteins A, D, and C1q in the clearance of apoptotic cells in vivo and in vitro: calreticulin and CD91 as a common collectin receptor complex. J Immunol. 2002;169:3978–86. doi: 10.4049/jimmunol.169.7.3978. [DOI] [PubMed] [Google Scholar]
  • 102.Hodge S, Hodge G, Scicchitano R, Reynolds PN, Holmes M. Alveolar macrophages from subjects with chronic obstructive pulmonary disease are deficient in their ability to phagocytose apoptotic airway epithelial cells. Immunol Cell Biol. 2003;81:289–96. doi: 10.1046/j.1440-1711.2003.t01-1-01170.x. [DOI] [PubMed] [Google Scholar]
  • 103.Yamaryo T, Oishi K, Yoshimine H, Tsuchihashi Y, Matsushima K, Nagatake T. Fourteen-member macrolides promote the phosphatidylserine receptor-dependent phagocytosis of apoptotic neutrophils by alveolar macrophages. Antimicrob Agents Chemother. 2003;47:48–53. doi: 10.1128/AAC.47.1.48-53.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hodge S, Hodge G, Brozyna S, Jersmann H, Holmes M, Reynolds PN. Azithromycin increases phagocytosis of apoptotic bronchial epithelial cells by alveolar macrophages. Eur Respir J. 2006;28:486–95. doi: 10.1183/09031936.06.00001506. [DOI] [PubMed] [Google Scholar]
  • 105.Hodge S, Hodge G, Ahern J, Jersmann H, Holmes M, Reynolds PN. Smoking alters alveolar macrophage recognition and phagocytic ability: Implications in COPD. Am J Respir Cell Mol Biol. 2007;37:748–755. doi: 10.1165/rcmb.2007-0025OC. [DOI] [PubMed] [Google Scholar]
  • 106.Monick MM, Carter AB, Gudmundsson G, Geist LJ, Hunninghake GW. Changes in PKC isoforms in human alveolar macrophages compared with blood monocytes. Am J Physiol. 1998;275:L389–97. doi: 10.1152/ajplung.1998.275.2.L389. [DOI] [PubMed] [Google Scholar]
  • 107.Magnus T, Chan A, Grauer O, Toyka KV, Gold R. Microglial phagocytosis of apoptotic inflammatory T cells leads to down-regulation of microglial immune activation. J Immunol. 2001;167:5004–10. doi: 10.4049/jimmunol.167.9.5004. [DOI] [PubMed] [Google Scholar]
  • 108.Stolzing A, Grune T. Neuronal apoptotic bodies: phagocytosis and degradation by primary microglial cells. FASEB J. 2004;18:743–5. doi: 10.1096/fj.03-0374fje. [DOI] [PubMed] [Google Scholar]
  • 109.Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3:23–35. doi: 10.1038/nri978. [DOI] [PubMed] [Google Scholar]
  • 110.Mosser DM. The many faces of macrophage activation. J Leukoc Biol. 2003;73:209–12. doi: 10.1189/jlb.0602325. [DOI] [PubMed] [Google Scholar]
  • 111.Edwards JP, Zhang X, Frauwirth KA, Mosser DM. Biochemical and functional characterization of three activated macrophage populations. J Leukoc Biol. 2006;80:1298–307. doi: 10.1189/jlb.0406249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Xu W, Roos A, Schlagwein N, Woltman AM, Daha MR, van Kooten C. IL-10-producing macrophages preferentially clear early apoptotic cells. Blood. 2006;107:4930–7. doi: 10.1182/blood-2005-10-4144. [DOI] [PubMed] [Google Scholar]
  • 113.Shortman K, Naik SH. Steady-state and inflammatory dendritic-cell development. Nat Rev Immunol. 2007;7:19–30. doi: 10.1038/nri1996. [DOI] [PubMed] [Google Scholar]
  • 114.Wu L, Liu YJ. Development of dendritic-cell lineages. Immunity. 2007;26:741–50. doi: 10.1016/j.immuni.2007.06.006. [DOI] [PubMed] [Google Scholar]
  • 115.Merad M, Manz MG, Karsunky H, Wagers A, Peters W, Charo I, Weissman IL, Cyster JG, Engleman EG. Langerhans cells renew in the skin throughout life under steady-state conditions. Nat Immunol. 2002;3:1135–41. doi: 10.1038/ni852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Sato N, Ahuja SK, Quinones M, Kostecki V, Reddick RL, Melby PC, Kuziel WA, Ahuja SS. CC chemokine receptor (CCR)2 is required for langerhans cell migration and localization of T helper cell type 1 (Th1)-inducing dendritic cells. Absence of CCR2 shifts the Leishmania major-resistant phenotype to a susceptible state dominated by Th2 cytokines, b cell outgrowth, and sustained neutrophilic inflammation. J Exp Med. 2000;192:205–18. doi: 10.1084/jem.192.2.205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Peters W, Cyster JG, Mack M, Schlondorff D, Wolf AJ, Ernst JD, Charo IF. CCR2-dependent trafficking of F4/80dim macrophages and CD11cdim/intermediate dendritic cells is crucial for T cell recruitment to lungs infected with Mycobacterium tuberculosis. J Immunol. 2004;172:7647–53. doi: 10.4049/jimmunol.172.12.7647. [DOI] [PubMed] [Google Scholar]
  • 118.Osterholzer JJ, Ames T, Polak T, Sonstein J, Moore BB, Chensue SW, Toews GB, Curtis JL. CCR2 and CCR6, but not endothelial selectins, mediate the accumulation of immature dendritic cells within the lungs of mice in response to particulate antigen. J Immunol. 2005;175:874–883. doi: 10.4049/jimmunol.175.2.874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Liu G, Wu C, Wu Y, Zhao Y. Phagocytosis of apoptotic cells and immune regulation. Scand J Immunol. 2006;64:1–9. doi: 10.1111/j.1365-3083.2006.01771.x. [DOI] [PubMed] [Google Scholar]
  • 120.Albert ML, Sauter B, Bhardwaj N. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs. Nature. 1998;392:86–9. doi: 10.1038/32183. [DOI] [PubMed] [Google Scholar]
  • 121.Albert ML, Pearce SF, Francisco LM, Sauter B, Roy P, Silverstein RL, Bhardwaj N. Immature dendritic cells phagocytose apoptotic cells via αvβ5 and CD36, and cross-present antigens to cytotoxic T lymphocytes. J Exp Med. 1998;188:1359–68. doi: 10.1084/jem.188.7.1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenous activators of dendritic cells. Nat Med. 1999;5:1249–1255. doi: 10.1038/15200. [DOI] [PubMed] [Google Scholar]
  • 123.Steinman RM, Turley S, Mellman I, Inaba K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med. 2000;191:411–6. doi: 10.1084/jem.191.3.411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Albert ML, Jegathesan M, Darnell RB. Dendritic cell maturation is required for the cross-tolerization of CD8+ T cells. Nat Immunol. 2001;2:1010–7. doi: 10.1038/ni722. [DOI] [PubMed] [Google Scholar]
  • 125.Stuart LM, Lucas M, Simpson C, Lamb J, Savill J, Lacy-Hulbert A. Inhibitory effects of apoptotic cell ingestion upon endotoxin-driven myeloid dendritic cell maturation. J Immunol. 2002;168:1627–1635. doi: 10.4049/jimmunol.168.4.1627. [DOI] [PubMed] [Google Scholar]
  • 126.Ip WK, Lau YL. Distinct maturation of, but not migration between, human monocyte-derived dendritic cells upon ingestion of apoptotic cells of early or late phases. J Immunol. 2004;173:189–96. doi: 10.4049/jimmunol.173.1.189. [DOI] [PubMed] [Google Scholar]
  • 127.Johansson U, Walther-Jallow L, Smed-Sorensen A, Spetz AL. Triggering of dendritic cell responses after exposure to activated, but not resting, apoptotic PBMCs. J Immunol. 2007;179:1711–20. doi: 10.4049/jimmunol.179.3.1711. [DOI] [PubMed] [Google Scholar]
  • 128.Rock KL, Hearn A, Chen CJ, Shi Y. Natural endogenous adjuvants. Springer Semin Immunopathol. 2005;26:231–46. doi: 10.1007/s00281-004-0173-3. [DOI] [PubMed] [Google Scholar]
  • 129.O'Keeffe M, Hochrein H, Vremec D, Caminschi I, Miller JL, Anders EM, Wu L, Lahoud MH, Henri S, Scott B, Hertzog P, Tatarczuch L, Shortman K. Mouse plasmacytoid cells: long-lived cells, heterogeneous in surface phenotype and function, that differentiate into CD8(+) dendritic cells only after microbial stimulus. J Exp Med. 2002;196:1307–19. doi: 10.1084/jem.20021031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Grouard G, Rissoan MC, Filgueira L, Durand I, Banchereau J, Liu YJ. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J Exp Med. 1997;185:1101–11. doi: 10.1084/jem.185.6.1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Liu YJ. IPC: Professional Type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu Rev Immunol. 2004;23:275–306. doi: 10.1146/annurev.immunol.23.021704.115633. [DOI] [PubMed] [Google Scholar]
  • 132.Dalgaard J, Beckstrom KJ, Jahnsen FL, Brinchmann JE. Differential capability for phagocytosis of apoptotic and necrotic leukemia cells by human peripheral blood dendritic cell subsets. J Leukoc Biol. 2005;77:689–698. doi: 10.1189/jlb.1204711. [DOI] [PubMed] [Google Scholar]
  • 133.Hoeffel G, Ripoche AC, Matheoud D, Nascimbeni M, Escriou N, Lebon P, Heshmati F, Guillet JG, Gannage M, Caillat-Zucman S, Casartelli N, Schwartz O, De la Salle H, Hanau D, Hosmalin A, Maranon C. Antigen crosspresentation by human plasmacytoid dendritic cells. Immunity. 2007;27:481–92. doi: 10.1016/j.immuni.2007.07.021. [DOI] [PubMed] [Google Scholar]
  • 134.Lu Q, Gore M, Zhang Q, Camenisch T, Boast S, Casagranda F, Lai C, Skinner MK, Klein R, Matsushima GK, Earp HS, Goff SP, Lemke G. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature. 1999;398:723–728. doi: 10.1038/19554. [DOI] [PubMed] [Google Scholar]
  • 135.O'Bryan JP, Frye RA, Cogswell PC, Neubauer A, Kitch B, Prokop C, Espinosa R. d., Le Beau MM, Earp HS, Liu ET. Axl, a transforming gene isolated from primary human myeloid leukemia cells, encodes a novel receptor tyrosine kinase. Mol Cell Biol. 1991;11:5016–31. doi: 10.1128/mcb.11.10.5016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Janssen JW, Schulz AS, Steenvoorden AC, Schmidberger M, Strehl S, Ambros PF, Bartram CR. A novel putative tyrosine kinase receptor with oncogenic potential. Oncogene. 1991;6:2113–20. [PubMed] [Google Scholar]
  • 137.Graham DK, Dawson TL, Mullaney DL, Snodgrass HR, Earp HS. Cloning and mRNA expression analysis of a novel human protooncogene, c-mer. Cell Growth Differ. 1994;5:647–667. [PubMed] [Google Scholar]
  • 138.Graham DK, Bowman GW, Dawson TL, Stanford WL, Earp HS, Snodgrass HR. Cloning and developmental expression analysis of the murine c-mer tyrosine kinase. Oncogene. 1995;10:2349–59. [PubMed] [Google Scholar]
  • 139.McCloskey P, Pierce J, Koski RA, Varnum B, Liu ET. Activation of the Axl receptor tyrosine kinase induces mitogenesis and transformation in 32D cells. Cell Growth Differ. 1994;5:1105–17. [PubMed] [Google Scholar]
  • 140.Sather S, Kenyon KD, Lefkowitz JB, Liang X, Varnum BC, Henson PM, Graham DK. A soluble form of the Mer receptor tyrosine kinase inhibits macrophage clearance of apoptotic cells and platelet aggregation. Blood. 2007;109:1026–33. doi: 10.1182/blood-2006-05-021634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Chen C, Li Q, Darrow AL, Wang Y, Derian CK, Yang J, de Garavilla L, Andrade-Gordon P, Damiano BP. Mer receptor tyrosine kinase signaling participates in platelet function. Arterioscler Thromb Vasc Biol. 2004;24:1118–23. doi: 10.1161/01.ATV.0000130662.30537.08. [DOI] [PubMed] [Google Scholar]
  • 142.Wang H, Chen S, Chen Y, Wang H, Wu H, Tang H, Xiong W, Ma J, Ge Y, Lu Q, Han D. The role of Tyro 3 subfamily receptors in the regulation of hemostasis and megakaryocytopoiesis. Haematologica. 2007;92:643–50. doi: 10.3324/haematol.10939. [DOI] [PubMed] [Google Scholar]
  • 143.Caraux A, Lu Q, Fernandez N, Riou S, Di Santo JP, Raulet DH, Lemke G, Roth C. Natural killer cell differentiation driven by Tyro3 receptor tyrosine kinases. Nat Immunol. 2006;7:747–54. doi: 10.1038/ni1353. [DOI] [PubMed] [Google Scholar]
  • 144.Ling L, Templeton D, Kung HJ. Identification of the major autophosphorylation sites of Nyk/Mer, an NCAM-related receptor tyrosine kinase. J Biol Chem. 1996;271:18355–18362. doi: 10.1074/jbc.271.31.18355. [DOI] [PubMed] [Google Scholar]
  • 145.Bellosta P, Costa M, Lin DA, Basilico C. The receptor tyrosine kinase ARK mediates cell aggregation by homophilic binding. Mol Cell Biol. 1995;15:614–25. doi: 10.1128/mcb.15.2.614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Budagian V, Bulanova E, Orinska Z, Thon L, Mamat U, Bellosta P, Basilico C, Adam D, Paus R, Bulfone-Paus S. A promiscuous liaison between IL-15 receptor and Axl receptor tyrosine kinase in cell death control. EMBO J. 2005;24:4260–70. doi: 10.1038/sj.emboj.7600874. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 147.Nagata K, Ohashi K, Nakano T, Arita H, Zong C, Hanafusa H, Mizuno K. Identification of the product of growth arrest-specific gene 6 as a common ligand for Axl, Sky, and Mer receptor tyrosine kinases. J Biol Chem. 1996;271:30022–7. doi: 10.1074/jbc.271.47.30022. [DOI] [PubMed] [Google Scholar]
  • 148.Stitt TN, Conn G, Gore M, Lai C, Bruno J, Radziejewski C, Mattsson K, Fisher J, Gies DR, Jones PF, Masiakowski P, Ryan TE, Tobkes NJ, Chen DH, DiStephano PS, Long GL, Basilico C, Goldfarb MP, Lemke G, Glass DJ, Yancopoulos GD. The anticoagulation factor protein S and its relative, Gas6, are ligands for the Tyro 3/Axl family of receptor tyrosine kinases. Cell. 1995;80:661–670. doi: 10.1016/0092-8674(95)90520-0. [DOI] [PubMed] [Google Scholar]
  • 149.Anderson HA, Maylock CA, Williams JA, Paweletz CP, Shu H, Shacter E. Serum-derived protein S binds to phosphatidylserine and stimulates the phagocytosis of apoptotic cells. Nat Immunol. 2003;4:87–91. doi: 10.1038/ni871. [DOI] [PubMed] [Google Scholar]
  • 150.Seitz HM, Camenisch TD, Lemke G, Earp HS, Matsushima GK. Macrophages and dendritic cells use different Axl/Mertk/Tyro3 receptors in clearance of apoptotic cells. J Immunol. 2007;178:5635–42. doi: 10.4049/jimmunol.178.9.5635. [DOI] [PubMed] [Google Scholar]
  • 151.Sasaki T, Knyazev PG, Clout NJ, Cheburkin Y, Gohring W, Ullrich A, Timpl R, Hohenester E. Structural basis for Gas6-Axl signalling. EMBO J. 2006;25:80–7. doi: 10.1038/sj.emboj.7600912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Budagian V, Bulanova E, Orinska Z, Duitman E, Brandt K, Ludwig A, Hartmann D, Lemke G, Saftig P, Bulfone-Paus S. Soluble Axl is generated by ADAM10-dependent cleavage and associates with Gas6 in mouse serum. Mol Cell Biol. 2005;25:9324–39. doi: 10.1128/MCB.25.21.9324-9339.2005. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 153.D'Cruz PM, Yasumura D, Weir J, Matthes MT, Abderrahim H, LaVail MM, Vollrath D. Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Hum Mol Genet. 2000;9:645–651. doi: 10.1093/hmg/9.4.645. [DOI] [PubMed] [Google Scholar]
  • 154.Gal A, Li Y, Thompson DA, Weir J, Orth U, Jacobson SG, Apfelstedt-Sylla E, Vollrath D. Mutations in MERTK, the human orthologue of the RCS rat retinal dystrophy gene, cause retinitis pigmentosa. Nat Genet. 2000;26:270–271. doi: 10.1038/81555. [DOI] [PubMed] [Google Scholar]
  • 155.Vollrath D, Feng W, Duncan JL, Yasumura D, D'Cruz PM, Chappelow A, Matthes MT, Kay MA, LaVail MM. Correction of the retinal dystrophy phenotype of the RCS rat by viral gene transfer of Mertk. Proc Natl Acad Sci U S A. 2001;98:12584–12589. doi: 10.1073/pnas.221364198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Lu Q, Lemke G. Homeostatic regulation of the immune system by receptor tyrosine kinases of the Tyro 3 family. Science. 2001;293:306–311. doi: 10.1126/science.1061663. [DOI] [PubMed] [Google Scholar]
  • 157.Scott RS, McMahon EJ, Pop SM, Reap EA, Caricchio R, Cohen PL, Earp HS, Matsushima GK. Phagocytosis and clearance of apoptotic cells is mediated by MER. Nature. 2001;411:207–211. doi: 10.1038/35075603. [DOI] [PubMed] [Google Scholar]
  • 158.Behrens EM, Gadue P, Gong SY, Garrett S, Stein PL, Cohen PL. The mer receptor tyrosine kinase: expression and function suggest a role in innate immunity. Eur J Immunol. 2003;33:2160–7. doi: 10.1002/eji.200324076. [DOI] [PubMed] [Google Scholar]
  • 159.Hu B, Punturieri A, Todt J, Sonstein J, Polak T, Curtis JL. Recognition and phagocytosis of apoptotic T cells by resident murine macrophages requires multiple signal transduction events. J Leukoc Biol. 2002;71:881–889. [PMC free article] [PubMed] [Google Scholar]
  • 160.Todt JC, Hu B, Curtis JL. The receptor tyrosine kinase MerTK activates phospholipase C gamma2 during recognition of apoptotic thymocytes by murine macrophages. J Leukoc Biol. 2004;75:705–713. doi: 10.1189/jlb.0903439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Larsen EC, DiGennaro JA, Saito N, Mehta S, Loegering DJ, Mazurkiewicz JE, Lennartz MR. Differential requirement for classic and novel PKC isoforms in respiratory burst and phagocytosis in RAW 264.7 cells. J Immunol. 2000;165:2809–17. doi: 10.4049/jimmunol.165.5.2809. [DOI] [PubMed] [Google Scholar]
  • 162.Holm A, Tejle K, Gunnarsson T, Magnusson KE, Descoteaux A, Rasmusson B. Role of protein kinase C alpha for uptake of unopsonized prey and phagosomal maturation in macrophages. Biochem Biophys Res Commun. 2003;302:653–8. doi: 10.1016/s0006-291x(03)00231-6. [DOI] [PubMed] [Google Scholar]
  • 163.Canetti C, Hu B, Curtis JL, Peters-Golden M. Syk activation is a leukotriene B4-regulated event involved in macrophage phagocytosis of IgG-coated targets but not apoptotic cells. Blood. 2003;102:1877–1883. doi: 10.1182/blood-2003-02-0534. [DOI] [PubMed] [Google Scholar]
  • 164.Greenberg S, Chang P, Wang DC, Xavier R, Seed B. Clustered syk tyrosine kinase domains trigger phagocytosis. Proc Natl Acad Sci U S A. 1996;93:1103–7. doi: 10.1073/pnas.93.3.1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Crowley MT, Costello PS, Fitzer-Attas CJ, Turner M, Meng F, Lowell C, Tybulewicz VL, DeFranco AL. A critical role for Syk in signal transduction and phagocytosis mediated by Fcgamma receptors on macrophages. J Exp Med. 1997;186:1027–39. doi: 10.1084/jem.186.7.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Huang Y, Wange RL. T cell receptor signaling: beyond complex complexes. J Biol Chem. 2004;279:28827–30. doi: 10.1074/jbc.R400012200. [DOI] [PubMed] [Google Scholar]
  • 167.Hoffmann PR, deCathelineau AM, Ogden CA, Leverrier Y, Bratton DL, Daleke DL, Ridley AJ, Fadok VA, Henson PM. Phosphatidylserine (PS) induces PS receptor-mediated macropinocytosis and promotes clearance of apoptotic cells. J Cell Biol. 2001;155:649–659. doi: 10.1083/jcb.200108080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ogden CA, deCathelineau A, Hoffmann PR, Bratton D, Ghebrehiwet B, Fadok VA, Henson PM. C1q and mannose binding lectin engagement of cell surface calreticulin and CD91 initiates macropinocytosis and uptake of apoptotic cells. J Exp Med. 2001;194:781–795. doi: 10.1084/jem.194.6.781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Giles KM, Hart SP, Haslett C, Rossi AG, Dransfield I. An appetite for apoptotic cells? Controversies and challenges. Br J Haematol. 2000;109:1–12. doi: 10.1046/j.1365-2141.2000.01805.x. [DOI] [PubMed] [Google Scholar]
  • 170.Krysko DV, Brouckaert G, Kalai M, Vandenabeele P, D'Herde K. Mechanisms of internalization of apoptotic and necrotic L929 cells by a macrophage cell line studied by electron microscopy. J Morphol. 2003;258:336–45. doi: 10.1002/jmor.10161. [DOI] [PubMed] [Google Scholar]
  • 171.Krysko DV, D'Herde K, Vandenabeele P. Clearance of apoptotic and necrotic cells and its immunological consequences. Apoptosis. 2006;11:1709–26. doi: 10.1007/s10495-006-9527-8. [DOI] [PubMed] [Google Scholar]
  • 172.deCathelineau AM, Henson PM. The final step in programmed cell death: phagocytes carry apoptotic cells to the grave. Essays Biochem. 2003;39:105–17. doi: 10.1042/bse0390105. [DOI] [PubMed] [Google Scholar]
  • 173.Ellis RE, Jacobson DM, Horvitz HR. Genes required for the engulfment of cell corpses during programmed cell death in Caenorhabditis elegans. Genetics. 1991;129:79–94. doi: 10.1093/genetics/129.1.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Jehle AW, Gardai SJ, Li S, Linsel-Nitschke P, Morimoto K, Janssen WJ, Vandivier RW, Wang N, Greenberg S, Dale BM, Qin C, Henson PM, Tall AR. ATP-binding cassette transporter A7 enhances phagocytosis of apoptotic cells and associated ERK signaling in macrophages. J Cell Biol. 2006;174:547–56. doi: 10.1083/jcb.200601030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Reddien PW, Horvitz HR. The engulfment process of programmed cell death in Caenorhabditis elegans. Annu Rev Cell Dev Biol. 2004;20:193–221. doi: 10.1146/annurev.cellbio.20.022003.114619. [DOI] [PubMed] [Google Scholar]
  • 176.Roos A, Xu W, Castellano G, Nauta AJ, Garred P, Daha MR, Van Kooten C. Mini-review: A pivotal role for innate immunity in the clearance of apoptotic cells. Eur J Immunol. 2004;34:921–9. doi: 10.1002/eji.200424904. [DOI] [PubMed] [Google Scholar]
  • 177.Tosello-Trampont AC, Kinchen JM, Brugnera E, Haney LB, Hengartner MO, Ravichandran KS. Identification of two signaling submodules within the CrkII/ELMO/Dock180 pathway regulating engulfment of apoptotic cells. Cell Death Differ. 2007;14:963–72. doi: 10.1038/sj.cdd.4402094. [DOI] [PubMed] [Google Scholar]
  • 178.Kinchen JM, Cabello J, Klingele D, Wong K, Feichtinger R, Schnabel H, Schnabel R, Hengartner MO. Two pathways converge at CED-10 to mediate actin rearrangement and corpse removal in C. elegans. Nature. 2005;434:93–9. doi: 10.1038/nature03263. [DOI] [PubMed] [Google Scholar]
  • 179.Nakaya M, Tanaka M, Okabe Y, Hanayama R, Nagata S. Opposite effects of rho family GTPases on engulfment of apoptotic cells by macrophages. J Biol Chem. 2006;281:8836–42. doi: 10.1074/jbc.M510972200. [DOI] [PubMed] [Google Scholar]
  • 180.deBakker CD, Haney LB, Kinchen JM, Grimsley C, Lu M, Klingele D, Hsu PK, Chou BK, Cheng LC, Blangy A, Sondek J, Hengartner MO, Wu YC, Ravichandran KS. Phagocytosis of apoptotic cells is regulated by a UNC-73/TRIO-MIG-2/RhoG signaling module and armadillo repeats of CED-12/ELMO. Curr Biol. 2004;14:2208–16. doi: 10.1016/j.cub.2004.12.029. [DOI] [PubMed] [Google Scholar]
  • 181.Albert ML, Kim JI, Birge RB. avb5 integrin recruits the CrkII-Dock180-rac1 complex for phagocytosis of apoptotic cells. Nat Cell Biol. 2000;2:899–905. doi: 10.1038/35046549. [DOI] [PubMed] [Google Scholar]
  • 182.Feng W, Yasumura D, Matthes MT, LaVail MM, Vollrath D. Mertk triggers uptake of photoreceptor outer segments during phagocytosis by cultured retinal pigment epithelial cells. J Biol Chem. 2002;277:17016–22. doi: 10.1074/jbc.M107876200. [DOI] [PubMed] [Google Scholar]
  • 183.Hubbard SR, Miller WT. Receptor tyrosine kinases: mechanisms of activation and signaling. Curr Opin Cell Biol. 2007;19:117–23. doi: 10.1016/j.ceb.2007.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Guttridge KL, Luft JC, Dawson TL, Kozlowska E, Mahajan NP, Varnum B, Earp HS. Mer receptor tyrosine kinase signaling: prevention of apoptosis and alteration of cytoskeletal architecture without stimulation or proliferation. J Biol Chem. 2002;277:24057–66. doi: 10.1074/jbc.M112086200. [DOI] [PubMed] [Google Scholar]
  • 185.Toshima J, Ohashi K, Iwashita S, Mizuno K. Autophosphorylation activity and association with Src family kinase of Sky receptor tyrosine kinase. Biochem Biophys Res Commun. 1995;209:656–663. doi: 10.1006/bbrc.1995.1549. [DOI] [PubMed] [Google Scholar]
  • 186.Lan Z, Wu H, Li W, Wu S, Lu L, Xu M, Dai W. Transforming activity of receptor tyrosine kinase tyro3 is mediated, at least in part, by the PI3 kinase-signaling pathway. Blood. 2000;95:633–638. [PubMed] [Google Scholar]
  • 187.Braunger J, Schleithoff L, Schulz AS, Kessler H, Lammers R, Ullrich A, Bartram CR, Janssen JW. Intracellular signaling of the Ufo/Axl receptor tyrosine kinase is mediated mainly by a multi-substrate docking-site. Oncogene. 1997;14:2619–31. doi: 10.1038/sj.onc.1201123. [DOI] [PubMed] [Google Scholar]
  • 188.Hafizi S, Alindri F, Karlsson R, Dahlback B. Interaction of Axl receptor tyrosine kinase with C1-TEN, a novel C1 domain-containing protein with homology to tensin. Biochem Biophys Res Commun. 2002;299:793–800. doi: 10.1016/s0006-291x(02)02718-3. [DOI] [PubMed] [Google Scholar]
  • 189.Mahajan NP, Earp HS. An SH2 domain-dependent, phosphotyrosine-independent interaction between Vav1 and the Mer receptor tyrosine kinase: a mechanism for localizing guanine nucleotide-exchange factor action. J Biol Chem. 2003;278:42596–603. doi: 10.1074/jbc.M305817200. [DOI] [PubMed] [Google Scholar]
  • 190.Ling L, Kung HJ. Mitogenic signals and transforming potential of Nyk, a newly identified neural cell adhesion molecule-related receptor tyrosine kinase. Mol Cell Biol. 1995;15:6582–92. doi: 10.1128/mcb.15.12.6582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Heiring C, Dahlback B, Muller YA. Ligand recognition and homophilic interactions in Tyro3: structural insights into the Axl/Tyro3 receptor tyrosine kinase family. J Biol Chem. 2004;279:6952–8. doi: 10.1074/jbc.M311750200. [DOI] [PubMed] [Google Scholar]
  • 192.Wu Y, Singh S, Georgescu MM, Birge RB. A role for Mer tyrosine kinase in alphavbeta5 integrin-mediated phagocytosis of apoptotic cells. J Cell Sci. 2005;118:539–53. doi: 10.1242/jcs.01632. [DOI] [PubMed] [Google Scholar]
  • 193.Nandrot EF, Anand M, Almeida D, Atabai K, Sheppard D, Finnemann SC. Essential role for MFG-E8 as ligand for alphavbeta5 integrin in diurnal retinal phagocytosis. Proc Natl Acad Sci U S A. 2007;104:12005–10. doi: 10.1073/pnas.0704756104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Verbovetski I, Bychkov H, Trahtemberg U, Shapira I, Hareuveni M, Ben-Tal O, Kutikov I, Gill O, Mevorach D. Opsonization of apoptotic cells by autologous iC3b facilitates clearance by immature dendritic cells, down-regulates DR and CD86, and up-regulates CC chemokine receptor 7. J Exp Med. 2002;196:1553–61. doi: 10.1084/jem.20020263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Sano H, Hsu DK, Apgar JR, Yu L, Sharma BB, Kuwabara I, Izui S, Liu FT. Critical role of galectin-3 in phagocytosis by macrophages. J Clin Invest. 2003;112:389–97. doi: 10.1172/JCI17592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Hisatomi T, Sakamoto T, Sonoda KH, Tsutsumi C, Qiao H, Enaida H, Yamanaka I, Kubota T, Ishibashi T, Kura S, Susin SA, Kroemer G. Clearance of apoptotic photoreceptors: elimination of apoptotic debris into the subretinal space and macrophage-mediated phagocytosis via phosphatidylserine receptor and integrin alphavbeta3. Am J Pathol. 2003;162:1869–79. doi: 10.1016/s0002-9440(10)64321-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Johnson JD, Hess KL, Cook-Mills JM. CD44, alpha(4) integrin, and fucoidin receptor-mediated phagocytosis of apoptotic leukocytes. J Leukoc Biol. 2003;74:810–20. doi: 10.1189/jlb.0303092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Devitt A, Parker KG, Ogden CA, Oldreive C, Clay MF, Melville LA, Bellamy CO, Lacy-Hulbert A, Gangloff SC, Goyert SM, Gregory CD. Persistence of apoptotic cells without autoimmune disease or inflammation in CD14−/− mice. J Cell Biol. 2004;167:1161–70. doi: 10.1083/jcb.200410057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Hart SP, Alexander KM, Dransfield I. Immune complexes bind preferentially to Fc gamma RIIA (CD32) on apoptotic neutrophils, leading to augmented phagocytosis by macrophages and release of proinflammatory cytokines. J Immunol. 2004;172:1882–7. doi: 10.4049/jimmunol.172.3.1882. [DOI] [PubMed] [Google Scholar]
  • 200.Norsworthy PJ, Fossati-Jimack L, Cortes-Hernandez J, Taylor PR, Bygrave AE, Thompson RD, Nourshargh S, Walport MJ, Botto M. Murine CD93 (C1qRp) contributes to the removal of apoptotic cells in vivo but is not required for C1q-mediated enhancement of phagocytosis. J Immunol. 2004;172:3406–14. doi: 10.4049/jimmunol.172.6.3406. [DOI] [PubMed] [Google Scholar]
  • 201.Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-Ullrich JE, Bratton DL, Oldenborg PA, Michalak M, Henson PM. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 2005;123:321–34. doi: 10.1016/j.cell.2005.08.032. [DOI] [PubMed] [Google Scholar]
  • 202.Stuart LM, Takahashi K, Shi L, Savill J, Ezekowitz RA. Mannose-binding lectin-deficient mice display defective apoptotic cell clearance but no autoimmune phenotype. J Immunol. 2005;174:3220–6. doi: 10.4049/jimmunol.174.6.3220. [DOI] [PubMed] [Google Scholar]
  • 203.Vachon E, Martin R, Plumb J, Kwok V, Vandivier RW, Glogauer M, Kapus A, Wang X, Chow CW, Grinstein S, Downey GP. CD44 is a phagocytic receptor. Blood. 2006;107:4149–58. doi: 10.1182/blood-2005-09-3808. [DOI] [PubMed] [Google Scholar]
  • 204.Moffatt OD, Devitt A, Bell ED, Simmons DL, Gregory CD. Macrophage recognition of ICAM-3 on apoptotic leukocytes. J Immunol. 1999;162:6800–10. [PubMed] [Google Scholar]
  • 205.Hart SP, Ross JA, Ross K, Haslett C, Dransfield I. Molecular characterization of the surface of apoptotic neutrophils: implications for functional downregulation and recognition by phagocytes. Cell Death Differ. 2000;7:493–503. doi: 10.1038/sj.cdd.4400680. [DOI] [PubMed] [Google Scholar]
  • 206.Gregory CD, Devitt A. The macrophage and the apoptotic cell: an innate immune interaction viewed simplistically? Immunology. 2004;113:1–14. doi: 10.1111/j.1365-2567.2004.01959.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Hanayama R, Tanaka M, Miyasaka K, Aozasa K, Koike M, Uchiyama Y, Nagata S. Autoimmune disease and impaired uptake of apoptotic cells in MFG-E8-deficient mice. Science. 2004;304:1147–50. doi: 10.1126/science.1094359. [DOI] [PubMed] [Google Scholar]
  • 208.Cohen PL, Caricchio R, Abraham V, Camenisch TD, Jennette JC, Roubey RA, Earp HS, Matsushima G, Reap EA. Delayed apoptotic cell clearance and lupus-like autoimmunity in mice lacking the c-mer membrane tyrosine kinase. J Exp Med. 2002;196:135–140. doi: 10.1084/jem.20012094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Hanayama R, Miyasaka K, Nakaya M, Nagata S. MFG-E8-dependent clearance of apoptotic cells, and autoimmunity caused by its failure. Curr Dir Autoimmun. 2006;9:162–72. doi: 10.1159/000090780. [DOI] [PubMed] [Google Scholar]
  • 210.Camenisch TD, Koller BH, Earp HS, Matsushima GK. A novel receptor tyrosine kinase, Mer, inhibits TNF-alpha production and lipopolysaccharide-induced endotoxic shock. J Immunol. 1999;162:3498–3503. [PubMed] [Google Scholar]
  • 211.Sen P, Wallet MA, Yi Z, Huang Y, Henderson M, Mathews CE, Earp HS, Matsushima G, Baldwin AS, Jr., Tisch RM. Apoptotic cells induce Mer tyrosine kinase-dependent blockade of NF-kappaB activation in dendritic cells. Blood. 2007;109:653–60. doi: 10.1182/blood-2006-04-017368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Shimojima M, Takada A, Ebihara H, Neumann G, Fujioka K, Irimura T, Jones S, Feldmann H, Kawaoka Y. Tyro3 family-mediated cell entry of Ebola and Marburg viruses. J Virol. 2006;80:10109–16. doi: 10.1128/JVI.01157-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Sharif MN, Sosic D, Rothlin CV, Kelly E, Lemke G, Olson EN, Ivashkiv LB. Twist mediates suppression of inflammation by type I IFNs and Axl. J Exp Med. 2006;203:1891–901. doi: 10.1084/jem.20051725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Sosic D, Richardson JA, Yu K, Ornitz DM, Olson EN. Twist regulates cytokine gene expression through a negative feedback loop that represses NF-kappaB activity. Cell. 2003;112:169–80. doi: 10.1016/s0092-8674(03)00002-3. [DOI] [PubMed] [Google Scholar]
  • 215.Li Y, Gerbod-Giannone MC, Seitz H, Cui D, Thorp E, Tall AR, Matsushima GK, Tabas I. Cholesterol-induced apoptotic macrophages elicit an inflammatory response in phagocytes, which is partially attenuated by the Mer receptor. J Biol Chem. 2006;281:6707–17. doi: 10.1074/jbc.M510579200. [DOI] [PubMed] [Google Scholar]

RESOURCES