Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 May 1.
Published in final edited form as: Immunol Rev. 2017 May;277(1):158–173. doi: 10.1111/imr.12537

The many ways tissue phagocytes respond to dying cells

J Magarian Blander 1
PMCID: PMC5721677  NIHMSID: NIHMS857525  PMID: 28462530

Summary

Apoptosis is an important component of normal tissue physiology, and the prompt removal of apoptotic cells is equally essential to avoid the undesirable consequences of their accumulation and disintegration. Professional phagocytes are highly specialized for engulfing apoptotic cells. The recent ability to track cells that have undergone apoptosis in situ has revealed a division of labor among the tissue resident phagocytes that sample them. Macrophages are uniquely programmed to process internalized apoptotic cell-derived fatty acids, cholesterol and nucleotides, as a reflection of their dominant role in clearing the bulk of apoptotic cells. Dendritic cells carry apoptotic cells to lymph nodes where they signal the emergence and expansion of highly suppressive regulatory CD4 T cells. A broad suppression of inflammation is executed through distinct phagocyte-specific mechanisms. A clever induction of negative regulatory nodes is notable in dendritic cells serving to simultaneously shut down multiple pathways of inflammation. Several of the genes and pathways modulated in phagocytes in response to apoptotic cells have been linked to chronic inflammatory and autoimmune diseases such as atherosclerosis, inflammatory bowel disease and systemic lupus erythematosus. Our collective understanding of old and new phagocyte functions after apoptotic cell phagocytosis demonstrates the enormity of ways to mediate immune suppression and enforce tissue homeostasis.

Keywords: Dendritic Cells, Monocytes/Macrophages, Autoimmunity, Toll-like Receptors/Pattern Recognition Receptors, Apoptosis/Autophagy, Phagocytosis

Introduction

Cell death is maintained at an equilibrium with cell division in healthy tissues, and is essential for maintaining constant tissue size and proper tissue function. The preferred mode of death under homeostatic conditions is apoptosis, being that this process is conducted neatly where cells shrink and disassemble their contents without damaging neighboring cells. Apoptotic cells express several signals that initiate their engulfment by tissue resident phagocytes. First, a cell undergoing apoptosis will release ‘find-me’ signals that are directly or indirectly produced through activity of the apoptosis executioner caspases, caspase-3 and caspase-7. They include the partial hydrolysis product of plasma membrane phosphatidylcholine, lysophosphatidylcholine, sphingosine-1-phosphate also known as lysosphingolipid, nucleoside triphosphates, as well as chemotactic factors such as fractalkine/CX3CL1 packaged into microparticles that are released upon apoptosis (reviewed in (1)). Once phagocytes are called in, the second step is identification of the apoptotic cell through ‘eat-me’ signals including exposure of phosphatidyl-serine (PS) at the outer leaflet of the apoptotic cell plasma membrane via the combined activities of plasma membrane flippases and scramblases (reviewed in (2)). In the third step, receptors that mediate apoptotic cell engulfment either bind directly to the exposed PS on apoptotic cells (Stabilin-2 (Stab2), Brain angiogenesis inhibitor (BAI1) and T cell immunoglobulin domain-containing 4 (TIM4)) or indirectly through bridge molecules (Milk fat globule epidermal growth factor (MFG-E8), Growth arrest-specific gene 6 (Gas6), protein S or complement component C1q. The TAM family of receptor tyrosine kinases, TYRO3, Axl and Mer, are an example of the latter receptors (36), whereby Mer can bind to either Gas6 or protein S and Axl is constitutively bound to Gas6 in tissue (7). Yet other receptors bind to apoptotic cells independently of PS and these include the phagocytic receptor CD36 which links macrophages to apoptotic cells via Thrombospondin-1 acting as a bridge molecule (8). Another example is the LDL-related receptor protein 1 (LRP1 or CD91), which recognizes Calreticulin present on the surface of apoptotic cells or C1q that opsonizes apoptotic cells (911). Similarly, the scavenger receptor SCARF1 (12) and the Immunoglobulin superfamily member leukocyte-associated Ig-like receptor 1 (LAIR1, CD305) (13) can both recognize C1q on opsonized apoptotic cells and mediates their phagocytosis.

Successful clearance of apoptotic cells is critical for the preservation of immune tolerance and prevention of autoimmune and chronic inflammatory diseases (14). The prototypical autoimmune disease where a definitive link to apoptotic cell clearance has been made is systemic lupus erythematosus (SLE) (15, 16). Failed clearance of apoptotic cells contributes to their accumulation leading to their death by secondary necrosis which releases inflammatory molecules that activate the innate immune system. Persisting apoptotic cells are the source of self-antigens that can now be presented to the immune system within the context of inflammation, and stimulate the activation of self-reactive T and B cells. Frequently found in SLE patients are anti-double-stranded (ds)DNA, anti-histone, anti-Smith, anti-SS-B/La, anti-ribosomes (Ro) and anti-ribonucleoprotein (RNP) autoantibodies (17, 18). Immune complexes comprised of autoantibody and autoantigen bind to Toll-like receptor (TLR) 7 and TLR9 (19) and trigger the production of interferon (IFN)α, a hallmark of SLE (reviewed in (14, 20)).

Tissue positioning of phagocytes at sites of frequent apoptosis

The orchestrated process of apoptotic cell clearance enables macrophages to locate apoptotic cells within tissues, accurately identify apoptotic cells from viable cells, and initiate their prompt engulfment without interrupting tissue function. For example, in the brain where long-range identification of apoptotic neurons is paramount such as not to disrupt neuronal synapses, the fractalkine receptor CX3CR1 is highly expressed on microglia, the resident macrophages in the brain, and these cells also express high levels of MFG-E8, BAI1, TIM4 and TAM receptors (2, 21, 22). Microglia are abundant within the dentate gyrus, the site of neurogenesis where large numbers of neural progenitors undergo apoptosis during differentiation into neurons (2325). Such strategic positioning of macrophages in locations of frequent apoptosis is notable in all tissues, and the location reflects developmental patterns and physiology distinct to each tissue (reviewed in (26)). In the testis, macrophages are located within the interstitium and along seminiferous tubules, but their numbers are highest where undifferentiated spermatogonia concentrate (27). Spermatogenesis is associated with apoptosis of a large proportion of spermatogenic cells (28), and while prompt phagocytosis of these cells can conceivably be conducted by testicular macrophages, this task has so far been assigned to specialized Sertoli cells. These somatic lineage cells nurse spermatogenesis and are located within the seminiferous epithelium alongside developing germ cells (28). Sertoli cells express all three TAM receptors whose signaling is crucial for clearance of apoptotic germ cells during spermatogenesis (5). Sertoli cells also rely on a signaling pathway downstream of BAI involving the cytoplasmic protein Elmo1, which functions with DOCK1 as a guanine nucleotide exchange factor for the GTPase Rac1 to promote actin cytoskeletal rearrangement and apoptotic cell phagocytosis (29). Testicular macrophages, on the other hand, express components of the colony-stimulating factor 1 and retinoic acid pathways to support spermatogonial differentiation (27). ‘Tingible body’ macrophages, a familiar term referring to macrophages containing apoptotic cells or apoptotic cell fragments, are most notable in germinal centers within secondary lymphoid organs after the peak of an immune response when almost 10% of plasma cells undergo apoptosis (30). Notably, the number of ‘tingible body’ macrophages is reduced in the germinal centers of several patients with SLE (31) reflecting the impaired clearance of apoptotic cells in this disease.

Despite the large numbers of cells that undergo apoptosis in tissues on a daily basis, especially in tissues such as the intestine where these numbers in humans approximate 1010 daily (32), visualization of apoptotic cell engulfment by phagocytes in situ has been difficult due to the prompt and efficient nature of the process. The heterogeneity of professional phagocytes such as macrophages and dendritic cells in tissues adds complexity to the problem. It has presented the questions of which phagocytes in tissues clear dying cells and whether distinct phagocyte subsets are specialized in apoptotic cell uptake in vivo. Several studies have resorted to the injection of fluorescently-labeled apoptotic cells into mice to follow their phagocytosis and its immune consequences. Intravenously injected apoptotic B cells or tumor cells were internalized by CD8α+ DC within the spleen and apoptotic cell antigen was cross-presented to antigen-specific CD8 T cells leading to their deletion as a mechanism of immune tolerance (33, 34). Within the pancreatic lymph nodes, CD11c+CD11b+CD8α DC were found carrying fluorescently-labeled dead cells that had been injected directly into the pancreas (35). Intra-pancreatic injection of dead β-islet cells promoted activation of β cell-reactive T cells, and this was also the case upon the induction of β cell death in vivo via injection of Streptozotocin, a cytotoxic chemical that β cells are sensitive to (35). Sub-capsular sinus macrophages lining the lymphatic endothelium overlying B cell follicles were found to capture subcutaneously injected apoptotic cells in an experimental setting (36). Within the lung parenchyma where dendritic cell (DC) subsets are in intimate contact with the bronchial and alveolar epithelium (37, 38), only the DC subset characterized by expression of the E-cadherin-binding integrin αE (CD103) captured intra-nasally delivered apoptotic thymocytes and trafficked these apoptotic cells to the draining lymph nodes for cross-presentation of apoptotic cell-derived antigens to CD8 T cells (36). The nature of phagocyte that recognizes, samples and/or internalizes apoptotic cells is likely dependent on the tissue and its physiological state at any given time.

Visualizing clearance of cells undergoing apoptosis in situ

Rapid turnover of intestinal epithelial cells (IEC) every 4–5 days marks the intestinal epithelium as a site of increased apoptosis during homeostatic conditions (32), and identifies it as an ideal setting for studying apoptotic cell clearance in vivo. Using a mouse model where expression of a transgene encoding an enhanced green fluorescent protein (eGFP) and diphtheria toxin receptor (DTR) fusion protein is directed specifically to IECs, the fate of IEC after apoptosis in situ could be visualized (39). The model is referred to as the VDTR mouse (39). The diphtheria toxin receptor enables the experimental induction of apoptosis as was determined by administering a carefully titrated concentration of diphtheria toxin that increased the levels of apoptosis over background (39). This leads to the sensitive capture of a larger number of phagocytes in the act of apoptotic IEC uptake, and importantly without causing inflammation or disrupting homeostatic conditions (39). The transgenic VDTR mouse model enabled visualization of phagocytes that had engulfed cells undergoing apoptosis in situ. The studies showed that within a heterogeneous network of intestinal lamina propria phagocytes (40, 41), both subsets of macrophages and a single CD103+ subset out of three DC subsets are responsible for apoptotic IEC sampling (39). These phagocytes are appropriately positioned for the task; facing the basolateral surface of IEC and capable of extending their dendrites through epithelial tight junctions to sample luminal contents, commensal bacteria (41, 42), and apoptotic IEC (39). After the administration of diphtheria toxin to VDTR mice, both macrophage subsets within the small intestinal lamina propria, identified as CD103+CD11b+ or CD103CD11b+, sampled the bulk of apoptotic IEC over time (39). Because the content of eGFP+ cargo identifies phagocytes that had sampled apoptotic IEC, the cells could be subjected to fluorescent activated cell sorting and their transcriptional profiles could then be compared to their eGFP devoid counterparts. A snapshot could be taken over time after the induction of apoptosis to determine the transcriptional changes in phagocytes upon apoptotic cell sampling (39). Analysis of the phagocyte transcriptomes revealed specialized responses, which will be considered next within the larger context of functions phagocytes are known to conduct in response to apoptotic cell sampling.

The first finding from studying the gene expression profiles of phagocytes bearing apoptotic cells was the distinct patterns of expression of apoptotic cell engulfment receptors by macrophages versus the CD103+ DC (39). The CD103+ DC upregulated the gene encoding CD205 which has been reported to detect apoptotic cell ligands (43), while macrophages upregulated genes involved in apoptotic cell clearance including genes encoding the scavenger receptor CD163, the bridge molecule Gas6 and the TAM receptor Mer (39). Other genes were also differentially expressed in macrophages versus CD103+ DC including upregulation of the genes encoding the fractalkine receptor CX3CR1, TIM4, Stabilin-1, and LRP1 in both macrophages subsets, Axl by CD103CD11b+ macrophages, and BAI1, Tyro3 and Stablin-2 by CD103+ DC. Differences in the expression of Axl and Mer have been noted in murine spleen and liver tissues as well as bone marrow derived macrophages (BMDM) and dendritic cells (BMDC). Axl expression is more abundant than Mer in BMDC and CD11c+ splenic DC, and vice versa, Mer is more abundant than Axl in BMDM and spleen CD68+ ‘tingible body’ macrophages (7). Tyro3 expression has been noted in BMDC and not BMDM (7). The reasons for these differential patterns of phagocytic receptor expression are presently unclear, but likely serve specialized phagocyte-specific functions in different anatomical sites and conditions.

Defective expression of engulfment receptors has been reported in patients with SLE (reviewed in (15, 44)). Deficiencies in the early components of complement, C1q and C4, are strongly associated with the development of SLE (reviewed in (45, 46)). This association is thought to reflect an impairment in opsonic C1q-mediated phagocytosis of apoptotic cells as well as C1q dependent induction of opsonins such as Gas6 and Protein S (44, 47). Low levels of protein S have been reported in a subset of SLE patients (48). A reduction in the expression of LAIR1 on B cells (49) and plasmacytoid DC (50) from patients with SLE has been reported. Some SLE patients were found to have high serum levels of MFG-E8 at concentrations that in vitro inhibit the phagocytosis of apoptotic cells, and may thus lead to similar effects in patients (51). High levels of circulating soluble MER have also been reported in SLE patients (52). When shed upon proteolytic cleavage by the metalloproteinase ADAM-17, the ectodomains of Axl and Mer serve as soluble decoy receptors that interfere with apoptotic cell engulfment (53, 54).

Suppression of inflammation and immune response by apoptotic cells

The well-appreciated immunosuppressive effects of apoptotic cells

One of the best-known consequences of apoptotic cell clearance is immunosuppression. The first evidence came decades ago from observations showing the failure of macrophages to release the pro-inflammatory mediator Thromboxane A2 upon ingestion of apoptotic neutrophils (55), and the ability of apoptotic cells to suppress monocyte production of pro-inflammatory cytokines and induce expression of the anti-inflammatory cytokine IL-10 in response to lipopolysaccharide (LPS) (56). Other studies at the time reported the resolution of acute inflammation by apoptotic cells in vivo (57, 58), and this was attributed to the increased levels of the immunosuppressive cytokine TGF-β (59, 60). Apoptotic cells themselves have been reported to produce IL-10 and TGF-β during the process of dying (61, 62). Several studies have demonstrated the induction of immune tolerance in different settings in response to DC phagocytosis of apoptotic cells (reviewed in (63, 64)). The therapeutic potential of apoptotic cell induced immunosuppression has been explored based on data in animal models demonstrating effective control of chronic inflammation in the settings of allograft rejection and graft-versus-host disease (65, 66), diabetes (67), arthritis (6871), septic shock (72), and other chronic and acute inflammatory conditions (reviewed in (73)). A recently completed phase I/IIa clinical trial concluded that a single infusion of early apoptotic donor cells to 13 patients with hematological malignancies a day prior to hematopoietic stem cell transplantation, is a safe and potentially effective prophylactic therapy for acute graft-versus-host disease (74). Other studies seeking to promote resolution in neutrophil-dominated inflammation in experimental systems, have devised strategies to induce neutrophil apoptosis in situ via pharmacological inhibition of cyclin-dependent kinases (75), flavones (76), or the tumor necrosis factor-related apoptosis inducing ligand (TRAIL) (77). These studies have noted promising results. It is important to note here that the induction of cell death is an important component of drug development efforts, but not all forms of cell death lead to immunosuppression. Achieving the desired anti-inflammatory effects of apoptotic cells is linked to their prompt and efficient clearance by professional phagocytes in order to prevent these cells from undergoing secondary necrosis, which is inflammatory (64). In other instances, such as cancer therapy and some infectious diseases, immunogenic cell death is desirable for mobilizing a protective anti-tumor immune response (reviewed in (78)). The nature of the immune response to dying cells, whether it is tolerance or immunity, is dependent on the signals expressed or released by these cells and the corresponding innate receptor signaling pathways engaged within the responding phagocytes (64).

The receptor systems mediating apoptotic cell-induced immunosuppression

At the forefront of receptors that temper inflammation in response to apoptotic cells are the macrophage and DC expressed TAM receptors, Axl, Tyro and Mer, which are not coincidentally also involved in mediating the phagocytosis of apoptotic cells (79, 80). In a negative feedback loop, the induction of Axl expression by type I IFNs downstream of TLR engagement leads to Axl binding to the type I IFN receptor, subsequent upregulation of the suppressor of cytokine signaling proteins SOCS1 and SOCS3, and termination of the inflammatory response (81). The expression of TAM receptors can also be induced by apoptotic cell recognition, as seen in intestinal lamina propria macrophages and CD103+ DC (39), presumably to prevent the initiation of immune responses. Mice deficient in all three members, Tyro, Axl, and Mer, develop chronic inflammation, enlarged secondary lymphoid organs, autoantibodies and systemic autoimmunity (82). The pathological phenotype observed is likely a combination of impaired apoptotic cell clearance and hyper-activation of B and T lymphocytes in secondary lymphoid organs.

Another set of receptors that have been associated with the immunosuppressive effects of apoptotic cells are the nuclear hormone receptors peroxisome-proliferator-activated receptor (PPAR) and liver x receptor (LXR). These receptors are additionally important for apoptotic cell clearance (8386). The first report implicating PPARγ in apoptotic cell-induced immunosuppression demonstrated its activation in response to apoptotic cell uptake, and its attenuation of reactive oxygen species production by inflammatory peritoneal macrophages (83). The homeostatic clearance of apoptotic neutrophils has been linked to LXR such that neutrophils accumulate within the blood and tissues of LXR-deficient mice (87, 88). Interestingly, LXR activation upon apoptotic neutrophil engulfment represses transcription of the inflammatory cytokine IL-23 (88), and in this manner controls the levels of neutrophils in the blood by targeting the upstream regulator of IL-17 and granulocyte-colony stimulating factor (G-CSF) that are important for granulopoiesis (89). Global or macrophage-specific deletion of PPARδ or deficiency in LXRα/β or retinoid X receptor (RXR)α, which heterodimerizes with LXR, leads to defects in apoptotic cell clearance in vivo and consequent increases in serum levels of autoantibodies with progressive lupus-like autoimmune disease (84, 86, 90). The phenotype of these mice is similar to that of mice with impaired apoptotic cell removal (91). Apoptotic cell uptake by PPARδ, RXRα or LXRα/β deficient macrophages is unable to suppress LPS-induced IL-12, TNF-α, or IL-1β transcription and synthesis, and fails to induce TGF-β and IL-10 synthesis demonstrating that PPARδ, RXRα and LXR contribute to the anti-inflammatory response induced by apoptotic cells (84, 86, 90).

Lastly, Nr4a1, a member of the Nr4a subfamily of nuclear receptors is induced in resident peritoneal macrophages in response to the phagocytosis of apoptotic cells in a PS-dependent manner (92). Nr4a1 expression interferes with NF-κB signaling to repress the expression of inflammatory cytokines such as IL-12 and maintain self-tolerance in the face of increased cell death in a model of experimental murine lupus (92).

Subset-specific pathways of suppressing inflammation

The studies tracking apoptotic IEC into small intestinal lamina propria phagocytes have shown that phagocytes adopt two prominent transcriptional signatures in response to apoptotic cells: the ‘downregulation of inflammatory genes’ and ‘suppression of the immune response’ (39). No upregulated genes were shared among these phagocytes, and only two genes were down-modulated by all three subsets (39): Plac8 implicated in clearance of certain microorganisms (93), and Itgb7 encoding integrin-β7 that mediates homing to gut-associated lymphoid tissue. Integrin-β7 is notably the target for a current IBD therapy with humanized monoclonal antibodies Vedolizumab and Etrolizumab (94). Different sets of genes are downregulated in DC versus macrophages, some of which have not previously been associated with the response to apoptotic cells. Genes encoding pattern recognition receptors Clec4a, Clec4b1, Cd209a, and Tlr2 are downregulated in CD103+CD11b+ and CD103CD11b+ macrophages (39). TLR2 mediates the innate response to fungal β-glucans and bacterial peptidoglycan, lipoproteins and lipoteichoic acid (95). Clec4a encodes DCIR, a C-type lectin receptor and member of the dendritic cell immunoreceptor (DCIR) family that was first described as an attachment factor for human immunodeficiency virus (HIV)-1 in DC (96). DCIR has also been reported to potentiate DC cross-presentation of antigen to CD8 T cells (97) and sustain type I IFN signaling (98). Notably, a polymorphism in DCIR, SNP rs2377422, is a genetic susceptibility factor for SLE and Sjogren’s syndrome (99), two autoimmune diseases where excessive apoptosis and impaired apoptotic cell clearance contribute to disease pathogenesis (15, 100). Clec4b1 encodes DCAR, a different member of the DCIR family that delivers antigens to the endocytic pathway in DC and enhances antigen-specific T cell responses (101). Cd209a encodes the C-type lectin CD209a, the murine homolog of human DC-specific ICAM-3-grabbing non-integrin DC-SIGN or CD209, which has been reported to induce the development of pathogenic T helper 17 cells driving severe hepatic granulomatous inflammation in a murine model of schistosomiasis (102). Intestinal lamina propria CD103+CD11b+ and CD103CD11b+ macrophages also downregulated Alox5ap encoding arachidonate 5-lipooxygenase (39), which facilitates inflammatory leukotriene biosynthesis (103). 5-lipooxygenase associates to lipid rafts of apoptotic cell membranes and presumably leads to the synthesis of apoptotic cell ligands that stimulate PPARγ in macrophages (85).

On the other hand, intestinal lamina propria CD103+ downregulated transcripts related to components of the inflammasome including Nlrp3, Casp1, and Il1a (104), as well as genes encoding MAPK/ERK signaling proteins including Lrrk2, Map3k4, and Fos. Leucine rich repeat kinase 2 (LRRK2), is a multifunctional protein kinase known to be a prominent contributor to familial Parkinson’s disease (105). Genome wide association studies have also identified LRRK2 as a major susceptibility gene for Crohn’s disease (106108). Notably, the Parkinson’s disease linked LRRK2(R1441G) mutation has been associated with increasing pro-inflammatory cytokine secretion (109, 110), and certain LRRK2 polymorphisms may function similarly in IBD. Other genes that overlapped with IBD susceptibility loci and were modulated upon apoptotic IEC sampling included Pfkfb3 encoding PFKFB3 (the inducible isoform of 6-phosphofructo-2-kinase, iPFK2) which was upregulated in apoptotic IEC bearing CD103+ DC (39). PFKFB3 is a PPARγ target (111, 112) involved in active PPARγ-dependent suppression of diet-induced intestinal inflammation (113). Curiously, IL12b encoding the p40 subunit shared by IL-12 and IL-23, in which single nucleotide polymorphisms are associated with Crohn’s disease in Korean populations (114), was upregulated in apoptotic IEC bearing intestinal lamina propria CD103+ DC (39).

Classical and alternative complement genes C1qa (encoding complement component C1qa) and Cfp (encoding complement factor Properdin), respectively (115), were downregulated in CD103+ DC (39). While this may be counterintuitive in light of the role that complement initiators and Properdin play in the recognition and opsonic phagocytosis of apoptotic cells (116), dampening activation of the complement cascade may supersede these functions while also attenuating NLRP3 inflammasome activation (117, 118). Additionally, a regulator of complement activation Cfh encoding complement factor H that attenuates late steps of complement activation, is upregulated in CD103CD11b+ macrophages (39). Factor H binds to apoptotic cells and when internalized catalyzes the generation of intracellular C3b, which opsonizes the apoptotic cell surface to promote clearance and aid in preventing autoimmunity (reviewed in (116)).

A DC-specific upregulation of negative immune response regulators

Of the phagocytes sampling apoptotic IEC, CD103+ DC upregulated genes involved in the negative regulation of inflammatory signaling (39). Notably, CD103+ DC appeared to be the only cells to do so. A common feature of the negative regulators is that they target more than one pathway of inflammation including the inflammasome, type I IFN, and MAPK signaling. This makes them ideal for enforcing immunosuppression at more than one level and in the face of more than one type of inflammatory stimulus. An example of a gene whose expression is induced in response to apoptotic cells is Nlrc5 (39), which encodes a member of the Nod-like receptor family NLRC5 and works to inhibit both NF-κB and type I IFN signaling (119). NLRC5 suppresses the activation of IKK by TLR ligands through blocking the assembly of a functional IKK complex by interacting directly with the IKK catalytic subunits, IKKα and IKKβ, to inhibit their phosphorylation and block their association with NEMO (119). NLRC5 also binds to the RIG-I like receptors, RIG-I and MDA-5, which are activated by double-stranded and single-stranded DNAs or certain viruses (120), and prevents their association with the mitochondrial anti-viral signaling protein MAVS and induction of a downstream type I IFN response (119).

Another example of a negative regulator that targets more than one pathway of inflammation is the protein A20 encoded by Tnfaip3, and whose expression is upregulated in small intestinal lamina propria CD103+ DC in response to apoptotic IEC (39). Mice deficient in A20 exhibit severe systemic inflammation, cachexia, increased sensitivity to LPS and TNF, and premature death (121). Deletion of A20 in specific cell types in mice leads to autoimmunity and organ-specific inflammation (122128) similar to those in humans identified to have polymorphisms and mutations in Tnfaip3 gene (129, 130). Deletion of A20 in DC leads to the development of pathologies in mice similar to those seen in humans with inflammatory bowel disease and SLE including colitis, ankylosing spondylitis, autoantibody development, nephritis, and splenomegaly (123, 125). A20 is a ubiquitin modifying enzyme that potently inhibits NF-κB signaling (131, 132), RIG-I signaling and antiviral responses (133, 134), as well as NLRP3 inflammasome activation (135, 136). A20 inhibits Nlrp3 transcription as a consequence of its inhibition of NF-κB signaling (135), but also exerts distinct effects on the function of the NLRP3 inflammasome complex by regulating its ubiquitination and composition (136). Enhanced apoptotic cell phagocytosis, presentation of apoptotic cell-derived antigen, and activation of self-reactive T cells has also been associated with A20 deficiency in DC (125). Notably, apoptotic cell uptake by A20 deficient DC fails to inhibit pro-inflammatory cytokine production in response to LPS (125). DC induction of A20 in response to apoptotic cells (39) may thus serve not only to inhibit inflammation, but also apoptotic cell antigen presentation. Combined with a shut down in apoptotic cell phagocytosis in a negative feedback loop (125), A20 might serve to limit the supply of self-antigen and its presentation within an inflammatory context.

Finally, CD103+ DC bearing apoptotic IEC show higher expression of Oasl1 transcripts compared to their devoid counterparts (39). Unlike the interferon-induced 2’-5’-oligoadenylate synthetases, the Oasl1 encoded interferon inducible oligoadenylate synthetase-like 1 (OASL1) is a non-enzymatic OAS protein that has two ubiquitin-like repeats (137). OASL1 inhibits the translation of IRF7, the master transcription factor for type I IFN, by binding to the 5’ untranslated region of the Irf7 mRNA (138). Apoptotic cell-carrying CD103+ DC also express higher levels of Spred1 (39), encoding for Spred1, which is highly expressed in hematopoietic cells (139) and belongs to the Sprouty (Spry) family of highly conserved proteins that modulate growth factor mediated mitogen-activated protein kinase activation (140, 141). The negative regulatory effects of Spred-1 have been shown in the context of allergen-induced airway eosinophilia and hyper-responsiveness as well as IL-3 signaling in hematopoietic cells (139, 142). Collectively, the transcriptional expression patterns discussed in this and the previous section reflect an overall genome-wide transcriptional suppression of inflammatory genes in response to apoptotic IEC sampling in situ.

Metabolic homeostasis in phagocytes post an apoptotic cell meal

When macrophages internalize apoptotic cells, they acquire the lipids, carbohydrates, protein, and nucleotides from the apoptotic cell. This content increases depending on the number of apoptotic cells a macrophage ingests, and poses a significant metabolic burden on the macrophage (143). Several studies have now shed light on the metabolic changes macrophages undergo to restore normal cellular function and homeostasis.

Increased lipid metabolism

Cholesterol derived from an engulfed apoptotic cell becomes incorporated into the cholesterol pool of engulfing macrophages increasing their cholesterol content by several fold (144). Several pieces of evidence show the mobilization of a homeostatic response aimed at restoring macrophage lipid metabolism. The increased levels of cholesterol in phagocytes following apoptotic cell phagocytosis had originally prompted a role for the fatty acid and sterol sensing nuclear receptors, PPAR and LXR, in apoptotic cell clearance. Apoptotic cell engulfment was found to induce the activation and expression of PPARγ (83, 145), PPARδ (90), and LXR (86) in macrophages. PPARs and LXRs form heterodimers with their obligate partners, the RXRs following binding to fatty acids and oxysterols, and recruit coactivators to induce transcription of a variety of genes involved in lipid and cholesterol metabolism (146, 147). Within the small intestinal lamina propria, increased expression of genes involved in lipid processing is notable in lamina propria resident macrophages that had engulfed apoptotic IEC (39). Intestinal CD103+CD11b+ macrophages upregulate the expression of Abca1, a direct target of LXR that encodes an ATP-binding cassette transporter ABCA1 also known as the cholesterol efflux regulatory protein (39). J774 macrophages and primary macrophages in tissue culture had similarly been reported to increase Abca1 transcription after the addition of apoptotic cells (144, 148). ABCA1 mediates the transport of cellular free cholesterol and phospholipid across the plasma membrane and transfers them primarily to circulating apolipoprotein A-I, leading to the formation of high density lipoprotein (HDL) particles (149). In turn, a small fraction of HDL, called Preβ-1 HDL, interacts with ABCA1 to remove cholesterol and phospholipids from cells (150). The exposure of PS on apoptotic cells is necessary and sufficient for both the upregulation of ABCA1 expression and the efflux of cholesterol (144). PS-dependent cholesterol efflux also requires the activity of PPARγ and LXR, suggesting communication between these nuclear receptors and PS-recognizing engulfment receptors (144). Evidence to date shows that signaling through the PS-specific phagocytic receptor BAI1 contributes to ABCA1 upregulation, albeit this was independent of LXR (148).

In macrophages, PPARγ controls the expression of CD36, transglutaminase-2, pentraxin-3, and Axl, all of which facilitate apoptotic cell phagocytosis (145, 151). PPARδ controls the expression of bridge molecules such as C1qb, Gas6, and Thrombospondin-1, and its deletion impairs the phagocytosis of apoptotic cells (90). Similarly, apoptotic cell clearance was reduced in macrophages deficient for both LXRα and LXRβ (86). Heterodimerization of RXRα with PPARγ or LXR also controls the expression of Mer (84, 86).

Apoptotic cell uptake induces macrophage expression of uncoupling protein 2 (UCP2) (152), a finding that reflects the ability of fatty acids, such as those derived from apoptotic cells, to enhance Ucp2 transcription through the activation of PPARs (153, 154) and the sterol regulatory element binding protein (SREBP) (154, 155). UCP2 is a mitochondrial anion carrier protein that uncouples oxidative phosphorylation from ATP production by dissipating protons across the inner mitochondrial membrane, and also decreases reactive oxygen species (ROS) produced by electron transport (154). Fatty acids also regulate the activity of UCP2 by increasing its proton conductance across inner mitochondrial membranes (156). By lowering mitochondrial membrane potential, which inversely controls engulfment capacity, UCP2 enables macrophages to internalize more than one apoptotic cell at a time (152).

An increased level of Acadsb transcripts encoding the short/branched chain specific acyl-CoA dehydrogenase (ACADSB, also known as 2-methylbutyryl-CoA dehydrogenase) has been noted in intestinal lamina propria CD103CD11b+ macrophages containing apoptotic IEC (39). This mitochondrial matrix enzyme is a member of the acyl-CoA dehydrogenase (ACAD) family of enzymes which catalyze the dehydrogenation of acyl-CoA esters as the first step in the metabolism of fatty acids (157). ACADSB has the greatest activity toward short branched chain acyl-CoA derivatives particularly 2-methylbutyryl-CoA in the isoleucine pathway, isobutyryl-CoA and 2-methylhexanoyl-CoA, as well as short straight chain acyl-CoAs such as isobutyryl-CoA and hexanoyl-CoA. Mutations in Acadsb are associated with a rare inherited metabolic disorder, 2-methylbutyrylglycinuria, which manifests in seizures and psychomotor delays from a defect in L-isoleucine catabolism (158160).

Breakdown of macrophage lipid metabolism – the case of atherosclerosis

The regulation of lipid metabolism in macrophages upon phagocytosis of apoptotic cells extends past the maintenance of cell-autonomous homeostasis and occupies center stage in atherosclerosis. Elevated plasma levels of low density lipoprotein cholesterol (LDL-C) remain a strong risk factor for atherosclerosis (161, 162). The retention of these particles within the intima of arteries along with the production of ROS, lipid peroxidation products and inflammatory mediators, leads to the development of atherosclerotic plaques (163). Here, foam cells – macrophages that have ingested large amounts of cholesteryl fatty acids – accumulate along with necrotic cells arising from the failed clearance of apoptotic macrophages. Over time, the arterial wall breaks down, and the resultant activation of pro-coagulant and pro-thrombotic pathways precipitates occlusive thrombus formation and myocardial infarction (163). Atherosclerosis is the culmination of interrelated events including defective resolution of the chronic inflammation triggered by lipoproteins retained in the subendothelium, defective egress of inflammatory cells and impaired clearance of apoptotic macrophages (163165).

Notably, several of the genes involved in regulating lipid metabolism in macrophages have been closely associated with atherosclerosis. Genetic deficiency for ABCA1 reduces the plasma levels of HDL cholesterol (HDL-C), one of the parameters associated with higher risk for cardiovascular disease (149, 150, 166). In mouse models of atherosclerosis, macrophage specific deletion of ABCA1 resulted in a large accumulation of cholesterol in macrophages, and exacerbated atherosclerosis in the advanced stage of disease (167). ABCA1 also protects macrophages from undergoing apoptosis after the engulfment of apoptotic cells, and it may do so in two ways. First, the efflux of cholesterol would protect macrophages from endoplasmic reticulum stress-induced apoptosis in response to the accumulated cholesterol (168). In fact, ABCA1 upregulation in response to apoptotic cell uptake might make macrophages resistant to the extreme conditions of cholesterol loading that they encounter within atherosclerotic lesions (169). Second, the nascent HDL formed through the activity of ABCA1 protects macrophages from the oxidative burst induced by the increased content of oxidized phospholipids derived from internalized apoptotic cells (170). Deficient ABCA1 function could thus contribute to increased numbers of apoptotic macrophages and resultant necrosis within atherosclerotic plaques. Consistent with these observations, transgenic overexpression of BAI1, which induces Abca1 transcription, within a hyperlipidemic genetic background of deficiency for the low-density lipoprotein receptor 1 (LDLR1) improved the HDL/low density lipoprotein (LDL) ratios of mice fed an atherogenic Western diet (148). Conversely, BAI1 deficiency under the same conditions resulted in significantly reduced levels of serum lipids including total cholesterol and HDL (148).

Genetic polymorphisms in UCP2 have been associated with several metabolic diseases including obesity, diabetes, and atherosclerosis (154). UCP2 has been linked to the regulation of the size of atherosclerotic plaques in mouse models (171, 172). This is likely a reflection of the increased levels of local ROS and ROS-driven lipid oxidation as well as the impaired clearance of apoptotic cells. In fact, deficiency in many of the apoptotic cell engulfment receptors or bridging molecule, such as transglutaminase-2, MFG-E8, Thrombospondin-1, complement C1q, Gas6 and pentraxin-3 (all targets for PPARs and LXR, see above) has identified specific roles for these molecules within advanced atherosclerotic lesions in mouse models (173178). Notably, deficiency in Axl restricted to the hematopoietic compartment of athero-prone mice did not affect plaque inflammation, apoptotic cell clearance or disease progression, and this did not appear to be due to compensatory Mer functions (179). LXR expression in macrophages is critical for the reduction of atherosclerosis and concordantly, ligand activation of LXR and PPAR reverses atherosclerosis in mouse models (180, 181). However, the efficacy of these agonists in preventing cardiovascular disease in humans remains to be seen (182).

Several studies have also elucidated an important function for Mer in atherosclerosis. As mentioned above, Mer is a prominent target of LXR and as such its expression is regulated by increased levels of macrophage fatty acids and cholesteryl esters – as would be the case for foam macrophages in atherosclerotic lesions – and increased Mer expression enhances the efficiency of apoptotic cell clearance. Accordingly, Mer deficiency in mice prone to atherogenesis increases the numbers of uncleared apoptotic cells and necrotic areas within atherosclerotic plaques (183, 184). Impaired Mer function upon proteolytic cleavage of its extracellular domain by metalloproteinase ADAM-17 generates soluble Mer, which competes with Gas6 for binding to Mer, and disables its functions in apoptotic cell clearance and suppression of inflammation (54, 185187). Expression of a cleavage resistant form of Mer in the hematopoietic compartment of atherogenesis-prone LDLR1 deficient mice fed an atherogenic high-fat diet, enhanced apoptotic cell clearance, decreased plaque necrosis, and increased resolution (188). Analysis of carotid endarterectomy samples revealed a strong positive correlation between soluble MER levels and both necrosis within lesions and the manifestation of an ischemic attack or stroke (188). CD68+ macrophages nearest to the necrotic cores of human carotid plaques exhibit highest ADAM17 and lowest surface MER expression (189).

The induction of regulatory CD4 T cells - a specialty of DC carrying apoptotic cells

Apoptotic cell-induced TGF-β and TREG cells

One of the major ways whereby innate recognition of apoptotic cells induces immune tolerance and suppresses inflammation is by creating the ideal conditions for the development of immunosuppressive regulatory CD4 T (TREG) cells. These cells are marked by expression of the transcription factor Foxp3 and function to control organ-specific autoimmune diseases by suppressing the activation of autoreactive T cells, presumably to self-antigens such as those derived from apoptotic cells (190, 191). Phagocytosis of apoptotic cells in vitro leads to the production of TGF-β, IL-10 and the vitamin A metabolite retinoic acid (60, 192198). These molecules provide the cytokine milieu for the differentiation of induced TREG cells (199205), which arise in the periphery and comprise a population distinct from natural TREG cells that develop in the thymus (206). One of the conserved non-coding DNA sequences (CNS1) within the Foxp3 promoter in TREG cells is responsive to TGF-β (207), contains Smad3 and retinoic acid receptor binding sites, and is required for differentiation of induced TREG cells specifically in the gut-associated lymphoid tissues and mesenteric lymph nodes (MLN) (208). Intestinal lamina propria and MLN CD103+ DC uniquely drive FOXP3+ TREG cell generation (203, 205, 209, 210). Intestinal lamina propria CD11b+F4/80+ macrophages have also been shown to induce FOXP3+ TREG cells in vitro (211). Colonic CD11c+CD11b+F4/80+ cells, thought to be CX3CR1+ macrophages, maintain FOXP3 expression as well as suppressive activity of TREG cells during colitis in vivo through the production of IL-10 (212).

Studies in vitro have shown that DC phagocytosis of apoptotic cells instructs naïve CD4 T cell differentiation into TREG cells (192, 198). DC expression of the αv integrin family of apoptotic cell receptors, αvβ3 and αvβ5, enables binding to and phagocytosis of apoptotic cells via bridge molecules such as Thrombospondin-1 and MFG-E8 (213). Intestinal CD103+ and not CD103 DC could induce TREG cell differentiation without exogenously added TGF-β (213, 214). Intestinal CD103+ DC express the αvβ8 integrin, which activates latent TGF-β and induces TREG cell generation (213, 215217). These studies strongly implicate apoptotic cell-stimulated TGF-β production in the generation of TREG cells. Evidence in vivo linking TREG cells to apoptotic cell engulfment comes from several studies. Myeloid specific deletion of the integrin αv or β8 decreases TREG cell numbers in the intestine (213, 218). The infusion of apoptotic cells into mice expands the numbers of inducible TREG cells (61, 219, 220), and this requires the presence of macrophages (61). Experimental induction of apoptosis within the intestinal epithelium of mice leads to an expansion of induced TREG cells in MLN (39). DC carrying apoptotic IEC have been noted in the MLN (39, 221, 222), suggesting that TREG cells induced by such DC could have specificity to epithelial cell derived self-antigens.

Specialization in TREG cell differentiation is restricted to apoptotic cell carrying DC

A comparison of the transcriptional profile of apoptotic cell containing DC versus macrophages within the small intestinal lamina propria reveals a distinct ‘regulatory CD4 T cell activation’ signature that is confined to CD103+ DC and not CD103+CD11b+ or CD103CD11b+ macrophages (39). These CD103+ DC upregulate Aldh1a2 (39), which catalyzes the conversion of retinal to retinoic acid to generate TREG cells in the intestine (199, 203, 204). Also upregulated is Lrrc32 (39), which encodes LRRC32 (also known as GARP). Expression of this protein has been studied on TREG cells where it associates with and retains latent TGF-β on the cell surface to promote TREG-mediated suppression (223). The expression of GARP by DC in response to apoptotic cell sampling (39) may also serve a similar role. Among the phagocytes that sample apoptotic IECs, highest expression of CCR7, the chemokine receptor that mediates DC migration to lymph nodes, is found on CD103+ DC that had sampled apoptotic cells (39), and these DC also have upregulated Cd40 (39) which encodes the costimulatory molecule CD40 whose interaction with CD40L regulates TREG cell numbers under homeostatic conditions (224, 225). Also upregulated by CD103+ DC in response to apoptotic cell sampling is RelB encoding the Rel/NF-κB transcription factor family member RelB that shows constitutively active κB-binding activity in lymphoid tissues under steady state conditions (226). Curiously, steady state migratory DC in peripheral lymph nodes characterized by the expression of CD40, nuclear RelB and surface latent TGF-β have been reported to transport peripheral self-antigens to the draining lymph nodes in a CCR7-dependent manner, present them to T cells and induce the differentiation of naïve CD4 T cells into TREG cells (227).

Small intestinal lamina propria CD103+ DC that have sampled apoptotic cells upregulate the suppressive co-stimulatory molecule Cd274 (Programmed death ligand PD-L1) (39), which synergizes with TGF-β to promote the instruction of TREG cell differentiation by DC, and whose expression on TREG cells has been reported to promote their differentiation and maintain their function by enhancing Foxp3 expression (228). Also upregulated by these CD103+ DC in response to apoptotic cell sampling are the chemokine encoding genes Ccl22 and Ccl17 that have been found to induce efficient chemotaxis of intestinal lamina propria TREG cells (229). Interestingly, CCL22 and CCL17 were reported to be selectively expressed by small intestinal lamina propria DCs at high levels, and that TREG cells were in close proximity to these DCs (229). The collective upregulation of these genes upon apoptotic IEC sampling by lamina propria CD103+ DC sets the stage for instructing differentiation of TREG cells (39), enabling DC to directly impact the generation of these critical mediators of intestinal tolerance and homeostasis (203).

Within the MLNs, migratory CD103+ DC containing apoptotic cells share a transcriptional profile similar to equivalent apoptotic cell carrying CD103+ DC in the small intestinal lamina propria (39). Several transcripts involved in TREG cell differentiation are additionally increased in migratory MLN CD103+ DC relative to equivalent MLN DC that do not contain apoptotic cells, including Ido1 and Il10 (39). IL-10 is a critical player in the induction of TREG cells by tolerogenic DC (230, 231). Ido1 encodes indoleamine 2,3 dioxygenase-1 (IDO-1) whose expression in cultured BMDC is induced by apoptotic cells (232, 233), and whose activity can be induced by TGF-β (234). Furthermore, the administration of apoptotic cells to mice induces the expression of IDO in splenic marginal zone macrophages (235). IDO-1 promotes immune tolerance through various mechanisms including the suppression of effector T cell responses and IL-6 production by DC, as well as the induction of TREG cell differentiation (235238). IDO-1 also suppresses IL-12 production and induces IL-10 and TGF-β production by macrophages exposed to apoptotic cells (239, 240). This is dependent on its downstream effector, the metabolic-stress sensing protein kinase GCN2 (240). Myeloid specific deletion of GCN2 in lupus-prone mice increases pathology and mortality while agonist-driven activation of GCN2 protects those mice from disease (235).

Compared to apoptotic cell carrying CD103+ DC in the small intestinal lamina propria, MLN CD103+ DCs carrying apoptotic IECs express approximately 4-fold lower levels of Itgb8, which converts TGF-β into an active state, while the levels of Tgfb1 transcripts are comparable (39). Migratory apoptotic cell carrying DC may thus already be primed to induce TREG cells upon arrival in the MLN. Within the MLN, T cell co-stimulatory receptors (CD274, CD40), and secreted signals (retinoic acid (via Aldh1a2), CCL22), are higher in apoptotic cell carrying CD103+ DC relative to CD103+ DC that do not contain apoptotic cells (39). Consistent with this profile, the apoptotic cell content of CD103+ DC is the determining factor in their ability to induce Foxp3+ TREG cells ex-vivo, and notably without added TGF-β (39).

Degradation of apoptotic cell DNA

Apoptotic cell chromosomal DNA is fragmented via cell autonomous and macrophage dependent mechanisms (241). DNASE1 facilitates the breakdown of chromatin during apoptosis, and DNASE-I deficient mice develop SLE-like syndrome with anti-nuclear antibodies, glomerular immune complex deposition and glomerulonephritis (242). Lower levels of DNASE-1 have been noted in SLE patients correlating with high antibody titers against nucleosomal antigens (242244). Within cells undergoing apoptosis, the fragmentation of DNA into nucleosomal units is the sole responsibility of the Caspase-activated DNAse (CAD or DFF-40), which under non-apoptotic conditions is kept in check by its specific inhibitor (ICAD or DFF-45) (245).

DNASE1L3, a circulating DNase produced primarily by macrophages and DC, digests the genomic DNA in apoptotic cell derived membrane-bound vesicles called microparticles (246). These microparticles are smaller than apoptotic bodies and can be found in the plasma of SLE patients bound to IgG to form a type of immune complex that may contribute to pathogenesis (247, 248). A highly basic C-terminal peptide in DNASE1L3 enables its association with liposomes encapsulating DNA (249), and this property of DNASE1L3 is thought to enable its digestion of surface-exposed chromatin exposed on microparticles thereby preventing autoantibody binding (246, 250). Dnase1l3-deficient mice develop antibodies to dsDNA and chromatin and exhibit features of SLE including glomerulonephritis (246).

Although apoptotic cell DNA is rapidly degraded by circulating DNASEs, DNA is bound to autoantibodies in SLE or the cationic antimicrobial peptide LL37 in psoriasis, an autoimmune disease of the skin (251, 252). These complexes protect the DNA from degradation and deliver it via endocytosis to endosomal compartments within DC where it engages TLR9 and signals downstream type I IFN and pro-inflammatory cytokine production (251254). In psoriasis, LL37 can similarly interact with self RNA and trigger TLR7 and TLR8 in human DC (255). DNA and RNA containing immune complexes internalized by autoreactive B cells via the B cell receptor also gain access to endosomal compartments within B cells where they engage TLR9 and TLR7, respectively, and stimulate B cell proliferation (256, 257) (and reviewed in (19, 258)).

After phagocytosis by macrophages, apoptotic cell DNA is degraded within lysosomes by lysosomal DNASE2 (259, 260). Mice deficient in CAD or ICAD do not exhibit features of autoimmunity because apoptotic cell DNA is degraded by lysosomal DNASE2 after macrophage phagocytosis of CAD-deficient apoptotic cells (245). Deletion of Dnase2 in mice is embryonic lethal due to constitutive production of interferon-β (260). Lethality can be rescued by crossing to a deficiency in type I IFN receptor (261), but as they age those mice develop chronic polyarthritis resembling human rheumatoid arthritis (260). This phenotype is a result of TNF-α production by macrophages unable to degrade apoptotic cell DNA (260). DNASE2 deficiency in both the hematopoietic and stromal compartment is required for the development of arthritis in these mice (262). Deletion of TLR3, TLR9 or both TLR signaling adaptors TRIF and MyD88, did not impact the lethality of DNASE2 deficient mice suggesting that the interferon response is mediated by a TLR-independent mechanism (263). Analysis of DNASE2 deficient mouse fetuses showed liver and spleen accumulation of cyclic dinucleotide [G(2’,5’)pA(3’,5’)p] (2’,3’-cGAMP), the product of the cytoplasmic DNA sensor cyclic GMP-AMP synthase (cGAS) (264). DNASE2 deficient mouse embryonic fibroblasts produced 2’,3’-cGAMP in a cGAS-dependent manner during apoptotic cell engulfment (264). These results suggest that undigested apoptotic cell DNA in lysosomes gains access to the cytoplasm of the engulfing cell to activate cGAS and type I IFN production. Indeed, mice doubly deficient for DNASE2 and the 2’,3’-cGAMP-responsive protein stimulator of interferon genes (STING) – downstream of cGAS – were rescued from lethality and polyarthritis even though macrophages from these mice could not digest engulfed apoptotic cell DNA (265). Cytoplasmic DNA sensors rather than TLR9 are involved in DNASE2 deficient mice because in the absence of DNASE2, the DNA fragments necessary to engage TLR9 are no longer generated (266). In cells undergoing apoptosis through the mitochondrial pathway, the activation of caspases 9, 3 and 7 suppresses the production of type I IFN production in response to released mitochondrial DNA, which would otherwise activate the cGAS-STING pathway (267, 268). This evidence collectively points to the cGAS/STING pathway as a major driver of type I IFN in response to undigested apoptotic cell derived mitochondrial and chromosomal DNA. However, it is important to add a distinction here. Autoantibodies to nuclear antigens persist in DNASE2 and STING double knock-out mice, but they are no longer detectable on a background deficient for Unc93b1, the ER-associated chaperone for TLR7 and TLR9 trafficking to endosomes (269, 270). Of note, these autoantibodies were not specific to dsDNA but to RNA-associated antigens perhaps reflecting the excessive accumulation of apoptotic cell debris containing RNA-associated autoantigens (270). Collectively, the data reveal that lingering undigested DNA from apoptotic cells triggers a STING-dependent type I IFN response and TLR-dependent autoantibody production.

Paradoxically, STING has also been identified as a negative regulator in systemic autoimmunity driven by TLR activation (271). The surprising evidence comes from the observation that STING deficiency in autoimmune prone MRL/lpr mice leads to the development of more severe disease and shorter lifespan (272). In line with these findings, STING deletion abrogates the hallmark apoptotic cell-induced TGF-β, IL-10 and IDO production by DC, and these DC produce pro-inflammatory IL-6 instead (273, 274). It will be important to decipher the mechanisms underlying the suppressive activities of STING in the context of autoimmunity and the response to apoptotic cells.

In conclusion – one more aspect of the biology of apoptotic cell clearance in tissues

The breadth of functions that phagocytes orchestrate in response to apoptotic cells is truly remarkable, as illustrated here by the plethora of homeostatic and pathological conditions impacted by each step in the process of recognizing, sampling and internalizing apoptotic cells. The scope is large and cannot be covered in a single review (please see other recent reviews (275278)). One final point to make pertains to the additional role that non-professional phagocytes, such as epithelial cells, play in the clearance of apoptotic cells. This has been documented in the respiratory, colonic, and post-weaning mammary epithelium (279283). To add yet another layer of complexity, macrophages also play a role in this setting. During phagocytosis of apoptotic cells or under inflammatory conditions, macrophages release insulin-like growth factor 1 (IGF-1), which binds to IGF-1 receptor (IGF1R) specifically expressed on epithelial cells and decreases their appetite for apoptotic cells (284). Macrophages also release microvesicles and through IGF-1 redirect epithelial cells towards internalizing microvesicles instead (284). The preferential uptake of microvesicles over apoptotic cells serves to suppress epithelial cell inflammatory responses, and accordingly the deletion of IGF1R in airway epithelial cells exacerbates airway inflammation (284). Macrophage-derived microvesicles may contain anti-inflammatory mediators such as SOCS1 and SOCS3 proteins that were reported to be present within microvesicles released by alveolar macrophages (285). These proteins attenuated JAK-STAT signaling in alveolar epithelial cells during smoking-induced lung inflammation (285). There are undoubtedly many more ways in which phagocytes suppress inflammation, mediate immune tolerance, and orchestrate tissue homeostasis.

Acknowledgments

J.M.B. is indebted to Ryan J. Cummings for his outstanding contributions. J.M.B. would like to thank her laboratory members for fruitful discussions. J.M.B. and her laboratory are supported by NIH grants AI127658, AI123284, DK072201 the Burroughs Wellcome Fund, and a Leukemia and Lymphoma Society Scholar Award.

Footnotes

J.M.B. does not have financial or personal relationships that could be viewed as presenting a potential conflict of interest.

References

  • 1.Medina CB, Ravichandran KS. Do not let death do us part: 'find-me' signals in communication between dying cells and the phagocytes. Cell Death Differ. 2016;23:979–989. doi: 10.1038/cdd.2016.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Nagata S, Suzuki J, Segawa K, Fujii T. Exposure of phosphatidylserine on the cell surface. Cell Death Differ. 2016;23:952–961. doi: 10.1038/cdd.2016.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Scott RS, McMahon EJ, Pop SM, et al. Phagocytosis and clearance of apoptotic cells is mediated by MER. Nature. 2001;411:207–211. doi: 10.1038/35075603. [DOI] [PubMed] [Google Scholar]
  • 4.Lemke G, Burstyn-Cohen T. TAM receptors and the clearance of apoptotic cells. Ann N Y Acad Sci. 2010;1209:23–29. doi: 10.1111/j.1749-6632.2010.05744.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lu Q, Gore M, Zhang Q, et al. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature. 1999;398:723–728. doi: 10.1038/19554. [DOI] [PubMed] [Google Scholar]
  • 6.Burstyn-Cohen T, Lew ED, Traves PG, Burrola PG, Hash JC, Lemke G. Genetic dissection of TAM receptor-ligand interaction in retinal pigment epithelial cell phagocytosis. Neuron. 2012;76:1123–1132. doi: 10.1016/j.neuron.2012.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zagorska A, Traves PG, Lew ED, Dransfield I, Lemke G. Diversification of TAM receptor tyrosine kinase function. Nat Immunol. 2014;15:920–928. doi: 10.1038/ni.2986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Febbraio M, Hajjar DP, Silverstein RL. CD36: a class B scavenger receptor involved in angiogenesis, atherosclerosis, inflammation, and lipid metabolism. J Clin Invest. 2001;108:785–791. doi: 10.1172/JCI14006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gardai SJ, McPhillips KA, Frasch SC, et al. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell. 2005;123:321–334. doi: 10.1016/j.cell.2005.08.032. [DOI] [PubMed] [Google Scholar]
  • 10.Ogden CA, deCathelineau A, Hoffmann PR, et al. 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]
  • 11.Gaipl US, Kuenkele S, Voll RE, et al. Complement binding is an early feature of necrotic and a rather late event during apoptotic cell death. Cell Death Differ. 2001;8:327–334. doi: 10.1038/sj.cdd.4400826. [DOI] [PubMed] [Google Scholar]
  • 12.Ramirez-Ortiz ZG, Pendergraft WF, 3rd, Prasad A, et al. The scavenger receptor SCARF1 mediates the clearance of apoptotic cells and prevents autoimmunity. Nat Immunol. 2013;14:917–926. doi: 10.1038/ni.2670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Son M, Santiago-Schwarz F, Al-Abed Y, Diamond B. C1q limits dendritic cell differentiation and activation by engaging LAIR-1. Proc Natl Acad Sci U S A. 2012;109:E3160–3167. doi: 10.1073/pnas.1212753109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mahajan A, Herrmann M, Munoz LE. Clearance Deficiency and Cell Death Pathways: A Model for the Pathogenesis of SLE. Front Immunol. 2016;7:35. doi: 10.3389/fimmu.2016.00035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Shao WH, Cohen PL. Disturbances of apoptotic cell clearance in systemic lupus erythematosus. Arthritis Res Ther. 2011;13:202. doi: 10.1186/ar3206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Herrmann M, Voll RE, Zoller OM, Hagenhofer M, Ponner BB, Kalden JR. Impaired phagocytosis of apoptotic cell material by monocyte-derived macrophages from patients with systemic lupus erythematosus. Arthritis Rheum. 1998;41:1241–1250. doi: 10.1002/1529-0131(199807)41:7<1241::AID-ART15>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
  • 17.Ippolito A, Wallace DJ, Gladman D, et al. Autoantibodies in systemic lupus erythematosus: comparison of historical and current assessment of seropositivity. Lupus. 2011;20:250–255. doi: 10.1177/0961203310385738. [DOI] [PubMed] [Google Scholar]
  • 18.Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011;365:2110–2121. doi: 10.1056/NEJMra1100359. [DOI] [PubMed] [Google Scholar]
  • 19.Marshak-Rothstein A. Toll-like receptors in systemic autoimmune disease. Nat Rev Immunol. 2006;6:823–835. doi: 10.1038/nri1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Obermoser G, Pascual V. The interferon-alpha signature of systemic lupus erythematosus. Lupus. 2010;19:1012–1019. doi: 10.1177/0961203310371161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fourgeaud L, Traves PG, Tufail Y, et al. TAM receptors regulate multiple features of microglial physiology. Nature. 2016;532:240–244. doi: 10.1038/nature17630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Liu Y, Yang X, Guo C, Nie P, Liu Y, Ma J. Essential role of MFG-E8 for phagocytic properties of microglial cells. PLoS One. 2013;8:e55754. doi: 10.1371/journal.pone.0055754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Luo C, Koyama R, Ikegaya Y. Microglia engulf viable newborn cells in the epileptic dentate gyrus. Glia. 2016;64:1508–1517. doi: 10.1002/glia.23018. [DOI] [PubMed] [Google Scholar]
  • 24.Paolicelli RC, Bolasco G, Pagani F, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333:1456–1458. doi: 10.1126/science.1202529. [DOI] [PubMed] [Google Scholar]
  • 25.Schafer DP, Lehrman EK, Kautzman AG, et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron. 2012;74:691–705. doi: 10.1016/j.neuron.2012.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Elliott MR, Ravichandran KS. The Dynamics of Apoptotic Cell Clearance. Dev Cell. 2016;38:147–160. doi: 10.1016/j.devcel.2016.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.DeFalco T, Potter SJ, Williams AV, Waller B, Kan MJ, Capel B. Macrophages Contribute to the Spermatogonial Niche in the Adult Testis. Cell Rep. 2015;12:1107–1119. doi: 10.1016/j.celrep.2015.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rato L, Alves MG, Socorro S, Duarte AI, Cavaco JE, Oliveira PF. Metabolic regulation is important for spermatogenesis. Nat Rev Urol. 2012;9:330–338. doi: 10.1038/nrurol.2012.77. [DOI] [PubMed] [Google Scholar]
  • 29.Elliott MR, Zheng S, Park D, et al. Unexpected requirement for ELMO1 in clearance of apoptotic germ cells in vivo. Nature. 2010;467:333–337. doi: 10.1038/nature09356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gray EE, Cyster JG. Lymph node macrophages. J Innate Immun. 2012;4:424–436. doi: 10.1159/000337007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Baumann I, Kolowos W, Voll RE, et al. Impaired uptake of apoptotic cells into tingible body macrophages in germinal centers of patients with systemic lupus erythematosus. Arthritis Rheum. 2002;46:191–201. doi: 10.1002/1529-0131(200201)46:1<191::AID-ART10027>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 32.Blander JM. Death in the intestinal epithelium-basic biology and implications for inflammatory bowel disease. FEBS J. 2016;283:2720–2730. doi: 10.1111/febs.13771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Liu K, Iyoda T, Saternus M, Kimura Y, Inaba K, Steinman RM. Immune tolerance after delivery of dying cells to dendritic cells in situ. J Exp Med. 2002;196:1091–1097. doi: 10.1084/jem.20021215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Iyoda T, Shimoyama S, Liu K, et al. The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivo. J Exp Med. 2002;195:1289–1302. doi: 10.1084/jem.20020161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Turley S, Poirot L, Hattori M, Benoist C, Mathis D. Physiological beta cell death triggers priming of self-reactive T cells by dendritic cells in a type-1 diabetes model. J Exp Med. 2003;198:1527–1537. doi: 10.1084/jem.20030966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Asano K, Nabeyama A, Miyake Y, et al. CD169-positive macrophages dominate antitumor immunity by crosspresenting dead cell-associated antigens. Immunity. 2011;34:85–95. doi: 10.1016/j.immuni.2010.12.011. [DOI] [PubMed] [Google Scholar]
  • 37.Condon TV, Sawyer RT, Fenton MJ, Riches DW. Lung dendritic cells at the innate-adaptive immune interface. J Leukoc Biol. 2011;90:883–895. doi: 10.1189/jlb.0311134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hammad H, Lambrecht BN. Dendritic cells and airway epithelial cells at the interface between innate and adaptive immune responses. Allergy. 2011;66:579–587. doi: 10.1111/j.1398-9995.2010.02528.x. [DOI] [PubMed] [Google Scholar]
  • 39.Cummings RJ, Barbet G, Bongers G, et al. Different tissue phagocytes sample apoptotic cells to direct distinct homeostasis programs. Nature. 2016;539:565–569. doi: 10.1038/nature20138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bekiaris V, Persson EK, Agace WW. Intestinal dendritic cells in the regulation of mucosal immunity. Immunol Rev. 2014;260:86–101. doi: 10.1111/imr.12194. [DOI] [PubMed] [Google Scholar]
  • 41.Gross M, Salame TM, Jung S. Guardians of the Gut - Murine Intestinal Macrophages and Dendritic Cells. Front Immunol. 2015;6:254. doi: 10.3389/fimmu.2015.00254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Rescigno M, Urbano M, Valzasina B, et al. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2001;2:361–367. doi: 10.1038/86373. [DOI] [PubMed] [Google Scholar]
  • 43.Shrimpton RE, Butler M, Morel AS, Eren E, Hue SS, Ritter MA. CD205 (DEC-205): a recognition receptor for apoptotic and necrotic self. Mol Immunol. 2009;46:1229–1239. doi: 10.1016/j.molimm.2008.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Colonna L, Lood C, Elkon KB. Beyond apoptosis in lupus. Curr Opin Rheumatol. 2014;26:459–466. doi: 10.1097/BOR.0000000000000083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Macedo AC, Isaac L. Systemic Lupus Erythematosus and Deficiencies of Early Components of the Complement Classical Pathway. Front Immunol. 2016;7:55. doi: 10.3389/fimmu.2016.00055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Manderson AP, Botto M, Walport MJ. The role of complement in the development of systemic lupus erythematosus. Annu Rev Immunol. 2004;22:431–456. doi: 10.1146/annurev.immunol.22.012703.104549. [DOI] [PubMed] [Google Scholar]
  • 47.Galvan MD, Foreman DB, Zeng E, Tan JC, Bohlson SS. Complement component C1q regulates macrophage expression of Mer tyrosine kinase to promote clearance of apoptotic cells. J Immunol. 2012;188:3716–3723. doi: 10.4049/jimmunol.1102920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Suh CH, Hilliard B, Li S, Merrill JT, Cohen PL. TAM receptor ligands in lupus: protein S but not Gas6 levels reflect disease activity in systemic lupus erythematosus. Arthritis Res Ther. 2010;12:R146. doi: 10.1186/ar3088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Colombo BM, Canevali P, Magnani O, et al. Defective expression and function of the leukocyte associated Ig-like receptor 1 in B lymphocytes from systemic lupus erythematosus patients. PLoS One. 2012;7:e31903. doi: 10.1371/journal.pone.0031903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kanakoudi-Tsakalidou F, Farmaki E, Tzimouli V, et al. Simultaneous changes in serum HMGB1 and IFN-alpha levels and in LAIR-1 expression on plasmatoid dendritic cells of patients with juvenile SLE. New therapeutic options? Lupus. 2014;23:305–312. doi: 10.1177/0961203313519157. [DOI] [PubMed] [Google Scholar]
  • 51.Yamaguchi H, Takagi J, Miyamae T, et al. Milk fat globule EGF factor 8 in the serum of human patients of systemic lupus erythematosus. J Leukoc Biol. 2008;83:1300–1307. doi: 10.1189/jlb.1107730. [DOI] [PubMed] [Google Scholar]
  • 52.Zizzo G, Guerrieri J, Dittman LM, Merrill JT, Cohen PL. Circulating levels of soluble MER in lupus reflect M2c activation of monocytes/macrophages, autoantibody specificities and disease activity. Arthritis Res Ther. 2013;15:R212. doi: 10.1186/ar4407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mullberg J, Rauch CT, Wolfson MF, et al. Further evidence for a common mechanism for shedding of cell surface proteins. FEBS Lett. 1997;401:235–238. doi: 10.1016/s0014-5793(96)01480-9. [DOI] [PubMed] [Google Scholar]
  • 54.Thorp E, Vaisar T, Subramanian M, Mautner L, Blobel C, Tabas I. Shedding of the Mer tyrosine kinase receptor is mediated by ADAM17 protein through a pathway involving reactive oxygen species, protein kinase Cdelta, and p38 mitogen-activated protein kinase (MAPK) J Biol Chem. 2011;286:33335–33344. doi: 10.1074/jbc.M111.263020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Meagher LC, Savill JS, Baker A, Fuller RW, Haslett C. Phagocytosis of apoptotic neutrophils does not induce macrophage release of thromboxane B2. J Leukoc Biol. 1992;52:269–273. [PubMed] [Google Scholar]
  • 56.Voll RE, Herrmann M, Roth EA, Stach C, Kalden JR, Girkontaite I. Immunosuppressive effects of apoptotic cells. Nature. 1997;390:350–351. doi: 10.1038/37022. [DOI] [PubMed] [Google Scholar]
  • 57.Cox G, Crossley J, Xing Z. Macrophage engulfment of apoptotic neutrophils contributes to the resolution of acute pulmonary inflammation in vivo. Am J Respir Cell Mol Biol. 1995;12:232–237. doi: 10.1165/ajrcmb.12.2.7865221. [DOI] [PubMed] [Google Scholar]
  • 58.Haslett C. Granulocyte apoptosis and its role in the resolution and control of lung inflammation. Am J Respir Crit Care Med. 1999;160:S5–11. doi: 10.1164/ajrccm.160.supplement_1.4. [DOI] [PubMed] [Google Scholar]
  • 59.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]
  • 60.Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998;101:890–898. doi: 10.1172/JCI1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kleinclauss F, Perruche S, Masson E, et al. Intravenous apoptotic spleen cell infusion induces a TGF-beta-dependent regulatory T-cell expansion. Cell Death Differ. 2006;13:41–52. doi: 10.1038/sj.cdd.4401699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gao Y, Herndon JM, Zhang H, Griffith TS, Ferguson TA. Antiinflammatory effects of CD95 ligand (FasL)-induced apoptosis. J Exp Med. 1998;188:887–896. doi: 10.1084/jem.188.5.887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Steinman RM, Turley S, Mellman I, Inaba K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med. 2000;191:411–416. doi: 10.1084/jem.191.3.411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol. 2009;9:353–363. doi: 10.1038/nri2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Morelli AE, Larregina AT. Apoptotic cell-based therapies against transplant rejection: role of recipient's dendritic cells. Apoptosis. 2010;15:1083–1097. doi: 10.1007/s10495-010-0469-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Morelli AE, Larregina AT. Concise Review: Mechanisms Behind Apoptotic Cell-Based Therapies Against Transplant Rejection and Graft versus Host Disease. Stem Cells. 2016;34:1142–1150. doi: 10.1002/stem.2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Xia CQ, Peng R, Qiu Y, Annamalai M, Gordon D, Clare-Salzler MJ. Transfusion of apoptotic beta-cells induces immune tolerance to beta-cell antigens and prevents type 1 diabetes in NOD mice. Diabetes. 2007;56:2116–2123. doi: 10.2337/db06-0825. [DOI] [PubMed] [Google Scholar]
  • 68.Bonnefoy F, Daoui A, Valmary-Degano S, Toussirot E, Saas P, Perruche S. Apoptotic cell infusion treats ongoing collagen-induced arthritis, even in the presence of methotrexate, and is synergic with anti-TNF therapy. Arthritis Res Ther. 2016;18:184. doi: 10.1186/s13075-016-1084-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gray M, Miles K, Salter D, Gray D, Savill J. Apoptotic cells protect mice from autoimmune inflammation by the induction of regulatory B cells. Proc Natl Acad Sci U S A. 2007;104:14080–14085. doi: 10.1073/pnas.0700326104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Notley CA, Brown MA, Wright GP, Ehrenstein MR. Natural IgM is required for suppression of inflammatory arthritis by apoptotic cells. J Immunol. 2011;186:4967–4972. doi: 10.4049/jimmunol.1003021. [DOI] [PubMed] [Google Scholar]
  • 71.Perruche S, Saas P, Chen W. Apoptotic cell-mediated suppression of streptococcal cell wall-induced arthritis is associated with alteration of macrophage function and local regulatory T-cell increase: a potential cell-based therapy? Arthritis Res Ther. 2009;11:R104. doi: 10.1186/ar2750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ren Y, Xie Y, Jiang G, et al. Apoptotic cells protect mice against lipopolysaccharide-induced shock. J Immunol. 2008;180:4978–4985. doi: 10.4049/jimmunol.180.7.4978. [DOI] [PubMed] [Google Scholar]
  • 73.Saas P, Daguindau E, Perruche S. Concise Review: Apoptotic Cell-Based Therapies-Rationale, Preclinical Results and Future Clinical Developments. Stem Cells. 2016;34:1464–1473. doi: 10.1002/stem.2361. [DOI] [PubMed] [Google Scholar]
  • 74.Mevorach D, Zuckerman T, Reiner I, et al. Single infusion of donor mononuclear early apoptotic cells as prophylaxis for graft-versus-host disease in myeloablative HLA-matched allogeneic bone marrow transplantation: a phase I/IIa clinical trial. Biol Blood Marrow Transplant. 2014;20:58–65. doi: 10.1016/j.bbmt.2013.10.010. [DOI] [PubMed] [Google Scholar]
  • 75.Rossi AG, Sawatzky DA, Walker A, et al. Cyclin-dependent kinase inhibitors enhance the resolution of inflammation by promoting inflammatory cell apoptosis. Nat Med. 2006;12:1056–1064. doi: 10.1038/nm1468. [DOI] [PubMed] [Google Scholar]
  • 76.Lucas CD, Allen KC, Dorward DA, et al. Flavones induce neutrophil apoptosis by down-regulation of Mcl-1 via a proteasomal-dependent pathway. FASEB J. 2013;27:1084–1094. doi: 10.1096/fj.12-218990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.McGrath EE, Marriott HM, Lawrie A, et al. TNF-related apoptosis-inducing ligand (TRAIL) regulates inflammatory neutrophil apoptosis and enhances resolution of inflammation. J Leukoc Biol. 2011;90:855–865. doi: 10.1189/jlb.0211062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17:97–111. doi: 10.1038/nri.2016.107. [DOI] [PubMed] [Google Scholar]
  • 79.Lemke G. Biology of the TAM receptors. Cold Spring Harb Perspect Biol. 2013;5:a009076. doi: 10.1101/cshperspect.a009076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Rothlin CV, Carrera-Silva EA, Bosurgi L, Ghosh S. TAM receptor signaling in immune homeostasis. Annu Rev Immunol. 2015;33:355–391. doi: 10.1146/annurev-immunol-032414-112103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rothlin CV, Ghosh S, Zuniga EI, Oldstone MB, Lemke G. TAM receptors are pleiotropic inhibitors of the innate immune response. Cell. 2007;131:1124–1136. doi: 10.1016/j.cell.2007.10.034. [DOI] [PubMed] [Google Scholar]
  • 82.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]
  • 83.Johann AM, von Knethen A, Lindemann D, Brune B. Recognition of apoptotic cells by macrophages activates the peroxisome proliferator-activated receptor-gamma and attenuates the oxidative burst. Cell Death Differ. 2006;13:1533–1540. doi: 10.1038/sj.cdd.4401832. [DOI] [PubMed] [Google Scholar]
  • 84.Roszer T, Menendez-Gutierrez MP, Lefterova MI, et al. Autoimmune kidney disease and impaired engulfment of apoptotic cells in mice with macrophage peroxisome proliferator-activated receptor gamma or retinoid X receptor alpha deficiency. J Immunol. 2011;186:621–631. doi: 10.4049/jimmunol.1002230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.von Knethen A, Sha LK, Kuchler L, et al. 5-Lipoxygenase contributes to PPARgamma activation in macrophages in response to apoptotic cells. Cell Signal. 2013;25:2762–2768. doi: 10.1016/j.cellsig.2013.08.045. [DOI] [PubMed] [Google Scholar]
  • 86.N AG, Bensinger SJ, Hong C, et al. Apoptotic cells promote their own clearance and immune tolerance through activation of the nuclear receptor LXR. Immunity. 2009;31:245–258. doi: 10.1016/j.immuni.2009.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Casanova-Acebes M, Pitaval C, Weiss LA, et al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell. 2013;153:1025–1035. doi: 10.1016/j.cell.2013.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hong C, Kidani Y, N AG, et al. Coordinate regulation of neutrophil homeostasis by liver X receptors in mice. J Clin Invest. 2012;122:337–347. doi: 10.1172/JCI58393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity. 2005;22:285–294. doi: 10.1016/j.immuni.2005.01.011. [DOI] [PubMed] [Google Scholar]
  • 90.Mukundan L, Odegaard JI, Morel CR, et al. PPAR-delta senses and orchestrates clearance of apoptotic cells to promote tolerance. Nat Med. 2009;15:1266–1272. doi: 10.1038/nm.2048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Nagata S, Hanayama R, Kawane K. Autoimmunity and the clearance of dead cells. Cell. 2010;140:619–630. doi: 10.1016/j.cell.2010.02.014. [DOI] [PubMed] [Google Scholar]
  • 92.Ipseiz N, Uderhardt S, Scholtysek C, et al. The nuclear receptor Nr4a1 mediates anti-inflammatory effects of apoptotic cells. J Immunol. 2014;192:4852–4858. doi: 10.4049/jimmunol.1303377. [DOI] [PubMed] [Google Scholar]
  • 93.Johnson RM, Kerr MS, Slaven JE. Plac8-dependent and inducible NO synthase-dependent mechanisms clear Chlamydia muridarum infections from the genital tract. Journal of immunology. 2012;188:1896–1904. doi: 10.4049/jimmunol.1102764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Mosli MH, Rivera-Nieves J, Feagan BG. T-cell trafficking and anti-adhesion strategies in inflammatory bowel disease: current and future prospects. Drugs. 2014;74:297–311. doi: 10.1007/s40265-013-0176-2. [DOI] [PubMed] [Google Scholar]
  • 95.Satoh T, Akira S. Toll-Like Receptor Signaling and Its Inducible Proteins. Microbiol Spectr. 2016;4 doi: 10.1128/microbiolspec.MCHD-0040-2016. [DOI] [PubMed] [Google Scholar]
  • 96.Lambert AA, Gilbert C, Richard M, Beaulieu AD, Tremblay MJ. The C-type lectin surface receptor DCIR acts as a new attachment factor for HIV-1 in dendritic cells and contributes to trans- and cis-infection pathways. Blood. 2008;112:1299–1307. doi: 10.1182/blood-2008-01-136473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Klechevsky E, Flamar AL, Cao Y, et al. Cross-priming CD8+ T cells by targeting antigens to human dendritic cells through DCIR. Blood. 2010;116:1685–1697. doi: 10.1182/blood-2010-01-264960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Troegeler A, Mercier I, Cougoule C, et al. C-type lectin receptor DCIR modulates immunity to tuberculosis by sustaining type I interferon signaling in dendritic cells. Proc Natl Acad Sci U S A. 2017 doi: 10.1073/pnas.1613254114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Liu M, Wu X, Liu X, et al. Contribution of dendritic cell immunoreceptor (DCIR) polymorphisms in susceptibility of systemic lupus erythematosus and primary Sjogren's syndrome. Hum Immunol. 2015;76:808–811. doi: 10.1016/j.humimm.2015.09.040. [DOI] [PubMed] [Google Scholar]
  • 100.Manganelli P, Fietta P. Apoptosis and Sjogren syndrome. Semin Arthritis Rheum. 2003;33:49–65. doi: 10.1053/sarh.2003.50019. [DOI] [PubMed] [Google Scholar]
  • 101.Kaden SA, Kurig S, Vasters K, et al. Enhanced dendritic cell-induced immune responses mediated by the novel C-type lectin receptor mDCAR1. J Immunol. 2009;183:5069–5078. doi: 10.4049/jimmunol.0900908. [DOI] [PubMed] [Google Scholar]
  • 102.Ponichtera HE, Shainheit MG, Liu BC, et al. CD209a expression on dendritic cells is critical for the development of pathogenic Th17 cell responses in murine schistosomiasis. J Immunol. 2014;192:4655–4665. doi: 10.4049/jimmunol.1400121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Radmark O, Werz O, Steinhilber D, Samuelsson B. 5-Lipoxygenase, a key enzyme for leukotriene biosynthesis in health and disease. Biochim Biophys Acta. 2015;1851:331–339. doi: 10.1016/j.bbalip.2014.08.012. [DOI] [PubMed] [Google Scholar]
  • 104.Blander JM. A long-awaited merger of the pathways mediating host defence and programmed cell death. Nature reviews Immunology. 2014;14:601–618. doi: 10.1038/nri3720. [DOI] [PubMed] [Google Scholar]
  • 105.Abeliovich A, Gitler AD. Defects in trafficking bridge Parkinson's disease pathology and genetics. Nature. 2016;539:207–216. doi: 10.1038/nature20414. [DOI] [PubMed] [Google Scholar]
  • 106.Anderson CA, Boucher G, Lees CW, et al. Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat Genet. 2011;43:246–252. doi: 10.1038/ng.764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Barrett JC, Hansoul S, Nicolae DL, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet. 2008;40:955–962. doi: 10.1038/NG.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Franke A, McGovern DP, Barrett JC, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci. Nat Genet. 2010;42:1118–1125. doi: 10.1038/ng.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Moehle MS, Webber PJ, Tse T, et al. LRRK2 inhibition attenuates microglial inflammatory responses. J Neurosci. 2012;32:1602–1611. doi: 10.1523/JNEUROSCI.5601-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Gillardon F, Schmid R, Draheim H. Parkinson's disease-linked leucine-rich repeat kinase 2(R1441G) mutation increases proinflammatory cytokine release from activated primary microglial cells and resultant neurotoxicity. Neuroscience. 2012;208:41–48. doi: 10.1016/j.neuroscience.2012.02.001. [DOI] [PubMed] [Google Scholar]
  • 111.Guo X, Xu K, Zhang J, et al. Involvement of inducible 6-phosphofructo-2-kinase in the anti-diabetic effect of peroxisome proliferator-activated receptor gamma activation in mice. J Biol Chem. 2010;285:23711–23720. doi: 10.1074/jbc.M110.123174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Nielsen R, Pedersen TA, Hagenbeek D, et al. Genome-wide profiling of PPARgamma:RXR and RNA polymerase II occupancy reveals temporal activation of distinct metabolic pathways and changes in RXR dimer composition during adipogenesis. Genes Dev. 2008;22:2953–2967. doi: 10.1101/gad.501108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Guo X, Li H, Xu H, et al. Disruption of inducible 6-phosphofructo-2-kinase impairs the suppressive effect of PPARgamma activation on diet-induced intestine inflammatory response. J Nutr Biochem. 2013;24:770–775. doi: 10.1016/j.jnutbio.2012.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Moon CM, Shin DJ, Son NH, et al. Genetic variants in the IL12B gene are associated with inflammatory bowel diseases in the Korean population. J Gastroenterol Hepatol. 2013;28:1588–1594. doi: 10.1111/jgh.12214. [DOI] [PubMed] [Google Scholar]
  • 115.Ghebrehiwet B, Hosszu KK, Valentino A, Peerschke EI. The C1q family of proteins: insights into the emerging non-traditional functions. Front Immunol. 2012;3 doi: 10.3389/fimmu.2012.00052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Martin M, Blom AM. Complement in removal of the dead - balancing inflammation. Immunol Rev. 2016;274:218–232. doi: 10.1111/imr.12462. [DOI] [PubMed] [Google Scholar]
  • 117.Arbore G, Kemper C. A novel 'Complement - Metabolism - Inflammasome Axis' as a key regulator of immune cell effector function. European journal of immunology. 2016 doi: 10.1002/eji.201546131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Benoit ME, Clarke EV, Morgado P, Fraser DA, Tenner AJ. Complement protein C1q directs macrophage polarization and limits inflammasome activity during the uptake of apoptotic cells. J Immunol. 2012;188:5682–5693. doi: 10.4049/jimmunol.1103760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Cui J, Zhu L, Xia X, et al. NLRC5 negatively regulates the NF-kappaB and type I interferon signaling pathways. Cell. 2010;141:483–496. doi: 10.1016/j.cell.2010.03.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Loo YM, Gale M., Jr Immune signaling by RIG-I-like receptors. Immunity. 2011;34:680–692. doi: 10.1016/j.immuni.2011.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lee EG, Boone DL, Chai S, et al. Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science. 2000;289:2350–2354. doi: 10.1126/science.289.5488.2350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chu Y, Vahl JC, Kumar D, et al. B cells lacking the tumor suppressor TNFAIP3/A20 display impaired differentiation and hyperactivation and cause inflammation and autoimmunity in aged mice. Blood. 2011;117:2227–2236. doi: 10.1182/blood-2010-09-306019. [DOI] [PubMed] [Google Scholar]
  • 123.Hammer GE, Turer EE, Taylor KE, et al. Expression of A20 by dendritic cells preserves immune homeostasis and prevents colitis and spondyloarthritis. Nat Immunol. 2011;12:1184–1193. doi: 10.1038/ni.2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Heger K, Fierens K, Vahl JC, et al. A20-deficient mast cells exacerbate inflammatory responses in vivo. PLoS Biol. 2014;12:e1001762. doi: 10.1371/journal.pbio.1001762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kool M, van Loo G, Waelput W, et al. The ubiquitin-editing protein A20 prevents dendritic cell activation, recognition of apoptotic cells, and systemic autoimmunity. Immunity. 2011;35:82–96. doi: 10.1016/j.immuni.2011.05.013. [DOI] [PubMed] [Google Scholar]
  • 126.Matmati M, Jacques P, Maelfait J, et al. A20 (TNFAIP3) deficiency in myeloid cells triggers erosive polyarthritis resembling rheumatoid arthritis. Nat Genet. 2011;43:908–912. doi: 10.1038/ng.874. [DOI] [PubMed] [Google Scholar]
  • 127.Tavares RM, Turer EE, Liu CL, et al. The ubiquitin modifying enzyme A20 restricts B cell survival and prevents autoimmunity. Immunity. 2010;33:181–191. doi: 10.1016/j.immuni.2010.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Vereecke L, Sze M, Mc Guire C, et al. Enterocyte-specific A20 deficiency sensitizes to tumor necrosis factor-induced toxicity and experimental colitis. J Exp Med. 2010;207:1513–1523. doi: 10.1084/jem.20092474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Mele A, Cervantes JR, Chien V, Friedman D, Ferran C. Single nucleotide polymorphisms at the TNFAIP3/A20 locus and susceptibility/resistance to inflammatory and autoimmune diseases. Adv Exp Med Biol. 2014;809:163–183. doi: 10.1007/978-1-4939-0398-6_10. [DOI] [PubMed] [Google Scholar]
  • 130.Zhou Q, Wang H, Schwartz DM, et al. Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet. 2016;48:67–73. doi: 10.1038/ng.3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Catrysse L, Vereecke L, Beyaert R, van Loo G. A20 in inflammation and autoimmunity. Trends Immunol. 2014;35:22–31. doi: 10.1016/j.it.2013.10.005. [DOI] [PubMed] [Google Scholar]
  • 132.Ma A, Malynn BA. A20: linking a complex regulator of ubiquitylation to immunity and human disease. Nat Rev Immunol. 2012;12:774–785. doi: 10.1038/nri3313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Feng W, Sun X, Shi N, Zhang M, Guan Z, Duan M. Influenza a virus NS1 protein induced A20 contributes to viral replication by suppressing interferon-induced antiviral response. Biochem Biophys Res Commun. 2017;482:1107–1113. doi: 10.1016/j.bbrc.2016.11.166. [DOI] [PubMed] [Google Scholar]
  • 134.Lin R, Yang L, Nakhaei P, et al. Negative regulation of the retinoic acid-inducible gene I-induced antiviral state by the ubiquitin-editing protein A20. J Biol Chem. 2006;281:2095–2103. doi: 10.1074/jbc.M510326200. [DOI] [PubMed] [Google Scholar]
  • 135.Vande Walle L, Van Opdenbosch N, Jacques P, et al. Negative regulation of the NLRP3 inflammasome by A20 protects against arthritis. Nature. 2014;512:69–73. doi: 10.1038/nature13322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Duong BH, Onizawa M, Oses-Prieto JA, et al. A20 restricts ubiquitination of pro-interleukin-1beta protein complexes and suppresses NLRP3 inflammasome activity. Immunity. 2015;42:55–67. doi: 10.1016/j.immuni.2014.12.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Eskildsen S, Justesen J, Schierup MH, Hartmann R. Characterization of the 2'-5'-oligoadenylate synthetase ubiquitin-like family. Nucleic Acids Res. 2003;31:3166–3173. doi: 10.1093/nar/gkg427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Lee MS, Kim B, Oh GT, Kim YJ. OASL1 inhibits translation of the type I interferon-regulating transcription factor IRF7. Nature immunology. 2013;14:346–355. doi: 10.1038/ni.2535. [DOI] [PubMed] [Google Scholar]
  • 139.Nonami A, Kato R, Taniguchi K, et al. Spred-1 negatively regulates interleukin-3-mediated ERK/mitogen-activated protein (MAP) kinase activation in hematopoietic cells. J Biol Chem. 2004;279:52543–52551. doi: 10.1074/jbc.M405189200. [DOI] [PubMed] [Google Scholar]
  • 140.Wakioka T, Sasaki A, Kato R, et al. Spred is a Sprouty-related suppressor of Ras signalling. Nature. 2001;412:647–651. doi: 10.1038/35088082. [DOI] [PubMed] [Google Scholar]
  • 141.Cabrita MA, Christofori G. Sprouty proteins, masterminds of receptor tyrosine kinase signaling. Angiogenesis. 2008;11:53–62. doi: 10.1007/s10456-008-9089-1. [DOI] [PubMed] [Google Scholar]
  • 142.Inoue H, Kato R, Fukuyama S, et al. Spred-1 negatively regulates allergen-induced airway eosinophilia and hyperresponsiveness. J Exp Med. 2005;201:73–82. doi: 10.1084/jem.20040616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Han CZ, Ravichandran KS. Metabolic connections during apoptotic cell engulfment. Cell. 2011;147:1442–1445. doi: 10.1016/j.cell.2011.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kiss RS, Elliott MR, Ma Z, Marcel YL, Ravichandran KS. Apoptotic cells induce a phosphatidylserine-dependent homeostatic response from phagocytes. Curr Biol. 2006;16:2252–2258. doi: 10.1016/j.cub.2006.09.043. [DOI] [PubMed] [Google Scholar]
  • 145.Majai G, Sarang Z, Csomos K, Zahuczky G, Fesus L. PPARgamma-dependent regulation of human macrophages in phagocytosis of apoptotic cells. Eur J Immunol. 2007;37:1343–1354. doi: 10.1002/eji.200636398. [DOI] [PubMed] [Google Scholar]
  • 146.Varga T, Czimmerer Z, Nagy L. PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim Biophys Acta. 2011;1812:1007–1022. doi: 10.1016/j.bbadis.2011.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Viswakarma N, Jia Y, Bai L, et al. Coactivators in PPAR-Regulated Gene Expression. PPAR Res. 2010;2010 doi: 10.1155/2010/250126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Fond AM, Lee CS, Schulman IG, Kiss RS, Ravichandran KS. Apoptotic cells trigger a membrane-initiated pathway to increase ABCA1. J Clin Invest. 2015;125:2748–2758. doi: 10.1172/JCI80300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Wang S, Smith JD. ABCA1 and nascent HDL biogenesis. Biofactors. 2014;40:547–554. doi: 10.1002/biof.1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Navab M, Reddy ST, Van Lenten BJ, Fogelman AM. HDL and cardiovascular disease: atherogenic and atheroprotective mechanisms. Nat Rev Cardiol. 2011;8:222–232. doi: 10.1038/nrcardio.2010.222. [DOI] [PubMed] [Google Scholar]
  • 151.Toth B, Garabuczi E, Sarang Z, et al. Transglutaminase 2 is needed for the formation of an efficient phagocyte portal in macrophages engulfing apoptotic cells. J Immunol. 2009;182:2084–2092. doi: 10.4049/jimmunol.0803444. [DOI] [PubMed] [Google Scholar]
  • 152.Park D, Han CZ, Elliott MR, et al. Continued clearance of apoptotic cells critically depends on the phagocyte Ucp2 protein. Nature. 2011;477:220–224. doi: 10.1038/nature10340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Kelly LJ, Vicario PP, Thompson GM, et al. Peroxisome proliferator-activated receptors gamma and alpha mediate in vivo regulation of uncoupling protein (UCP-1, UCP-2, UCP-3) gene expression. Endocrinology. 1998;139:4920–4927. doi: 10.1210/endo.139.12.6384. [DOI] [PubMed] [Google Scholar]
  • 154.Donadelli M, Dando I, Fiorini C, Palmieri M. UCP2, a mitochondrial protein regulated at multiple levels. Cell Mol Life Sci. 2014;71:1171–1190. doi: 10.1007/s00018-013-1407-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Medvedev AV, Robidoux J, Bai X, et al. Regulation of the uncoupling protein-2 gene in INS-1 beta-cells by oleic acid. J Biol Chem. 2002;277:42639–42644. doi: 10.1074/jbc.M208645200. [DOI] [PubMed] [Google Scholar]
  • 156.Beck V, Jaburek M, Demina T, et al. Polyunsaturated fatty acids activate human uncoupling proteins 1 and 2 in planar lipid bilayers. FASEB J. 2007;21:1137–1144. doi: 10.1096/fj.06-7489com. [DOI] [PubMed] [Google Scholar]
  • 157.Hiltunen JK, Qin Y. beta-oxidation - strategies for the metabolism of a wide variety of acyl-CoA esters. Biochim Biophys Acta. 2000;1484:117–128. doi: 10.1016/s1388-1981(00)00013-5. [DOI] [PubMed] [Google Scholar]
  • 158.Alfardan J, Mohsen AW, Copeland S, et al. Characterization of new ACADSB gene sequence mutations and clinical implications in patients with 2-methylbutyrylglycinuria identified by newborn screening. Mol Genet Metab. 2010;100:333–338. doi: 10.1016/j.ymgme.2010.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Gibson KM, Burlingame TG, Hogema B, et al. 2-Methylbutyryl-coenzyme A dehydrogenase deficiency: a new inborn error of L-isoleucine metabolism. Pediatr Res. 2000;47:830–833. doi: 10.1203/00006450-200006000-00025. [DOI] [PubMed] [Google Scholar]
  • 160.Madsen PP, Kibaek M, Roca X, et al. Short/branched-chain acyl-CoA dehydrogenase deficiency due to an IVS3+3A>G mutation that causes exon skipping. Hum Genet. 2006;118:680–690. doi: 10.1007/s00439-005-0070-4. [DOI] [PubMed] [Google Scholar]
  • 161.Shapiro MD, Fazio S. From Lipids to Inflammation: New Approaches to Reducing Atherosclerotic Risk. Circ Res. 2016;118:732–749. doi: 10.1161/CIRCRESAHA.115.306471. [DOI] [PubMed] [Google Scholar]
  • 162.Thomas MR, Lip GY. Novel Risk Markers and Risk Assessments for Cardiovascular Disease. Circ Res. 2017;120:133–149. doi: 10.1161/CIRCRESAHA.116.309955. [DOI] [PubMed] [Google Scholar]
  • 163.Tabas I, Garcia-Cardena G, Owens GK. Recent insights into the cellular biology of atherosclerosis. J Cell Biol. 2015;209:13–22. doi: 10.1083/jcb.201412052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Schrijvers DM, De Meyer GR, Herman AG, Martinet W. Phagocytosis in atherosclerosis: Molecular mechanisms and implications for plaque progression and stability. Cardiovasc Res. 2007;73:470–480. doi: 10.1016/j.cardiores.2006.09.005. [DOI] [PubMed] [Google Scholar]
  • 165.Viola J, Soehnlein O. Atherosclerosis - A matter of unresolved inflammation. Semin Immunol. 2015;27:184–193. doi: 10.1016/j.smim.2015.03.013. [DOI] [PubMed] [Google Scholar]
  • 166.Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis. 2016;59:97–106. doi: 10.1016/j.pcad.2016.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Bi X, Zhu X, Gao C, et al. Myeloid cell-specific ATP-binding cassette transporter A1 deletion has minimal impact on atherogenesis in atherogenic diet-fed low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol. 2014;34:1888–1899. doi: 10.1161/ATVBAHA.114.303791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Feng B, Yao PM, Li Y, et al. The endoplasmic reticulum is the site of cholesterol-induced cytotoxicity in macrophages. Nat Cell Biol. 2003;5:781–792. doi: 10.1038/ncb1035. [DOI] [PubMed] [Google Scholar]
  • 169.Cui D, Thorp E, Li Y, et al. Pivotal advance: macrophages become resistant to cholesterol-induced death after phagocytosis of apoptotic cells. J Leukoc Biol. 2007;82:1040–1050. doi: 10.1189/jlb.0307192. [DOI] [PubMed] [Google Scholar]
  • 170.Yvan-Charvet L, Pagler TA, Seimon TA, et al. ABCA1 and ABCG1 protect against oxidative stress-induced macrophage apoptosis during efferocytosis. Circ Res. 2010;106:1861–1869. doi: 10.1161/CIRCRESAHA.110.217281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Blanc J, Alves-Guerra MC, Esposito B, et al. Protective role of uncoupling protein 2 in atherosclerosis. Circulation. 2003;107:388–390. doi: 10.1161/01.cir.0000051722.66074.60. [DOI] [PubMed] [Google Scholar]
  • 172.Moukdar F, Robidoux J, Lyght O, Pi J, Daniel KW, Collins S. Reduced antioxidant capacity and diet-induced atherosclerosis in uncoupling protein-2-deficient mice. J Lipid Res. 2009;50:59–70. doi: 10.1194/jlr.M800273-JLR200. [DOI] [PubMed] [Google Scholar]
  • 173.Boisvert WA, Rose DM, Boullier A, et al. Leukocyte transglutaminase 2 expression limits atherosclerotic lesion size. Arterioscler Thromb Vasc Biol. 2006;26:563–569. doi: 10.1161/01.ATV.0000203503.82693.c1. [DOI] [PubMed] [Google Scholar]
  • 174.Ait-Oufella H, Kinugawa K, Zoll J, et al. Lactadherin deficiency leads to apoptotic cell accumulation and accelerated atherosclerosis in mice. Circulation. 2007;115:2168–2177. doi: 10.1161/CIRCULATIONAHA.106.662080. [DOI] [PubMed] [Google Scholar]
  • 175.Moura R, Tjwa M, Vandervoort P, Van Kerckhoven S, Holvoet P, Hoylaerts MF. Thrombospondin-1 deficiency accelerates atherosclerotic plaque maturation in ApoE−/− mice. Circ Res. 2008;103:1181–1189. doi: 10.1161/CIRCRESAHA.108.185645. [DOI] [PubMed] [Google Scholar]
  • 176.Norata GD, Marchesi P, Pulakazhi Venu VK, et al. Deficiency of the long pentraxin PTX3 promotes vascular inflammation and atherosclerosis. Circulation. 2009;120:699–708. doi: 10.1161/CIRCULATIONAHA.108.806547. [DOI] [PubMed] [Google Scholar]
  • 177.Lewis MJ, Malik TH, Ehrenstein MR, Boyle JJ, Botto M, Haskard DO. Immunoglobulin M is required for protection against atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation. 2009;120:417–426. doi: 10.1161/CIRCULATIONAHA.109.868158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Lutgens E, Tjwa M, Garcia de Frutos P, et al. Genetic loss of Gas6 induces plaque stability in experimental atherosclerosis. J Pathol. 2008;216:55–63. doi: 10.1002/path.2381. [DOI] [PubMed] [Google Scholar]
  • 179.Subramanian M, Proto JD, Matsushima GK, Tabas I. Deficiency of AXL in Bone Marrow-Derived Cells Does Not Affect Advanced Atherosclerotic Lesion Progression. Sci Rep. 2016;6:39111. doi: 10.1038/srep39111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Im SS, Osborne TF. Liver x receptors in atherosclerosis and inflammation. Circ Res. 2011;108:996–1001. doi: 10.1161/CIRCRESAHA.110.226878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Chinetti-Gbaguidi G, Staels B. PPARbeta in macrophages and atherosclerosis. Biochimie. 2016 doi: 10.1016/j.biochi.2016.12.008. [DOI] [PubMed] [Google Scholar]
  • 182.Kingwell BA, Chapman MJ, Kontush A, Miller NE. HDL-targeted therapies: progress, failures and future. Nat Rev Drug Discov. 2014;13:445–464. doi: 10.1038/nrd4279. [DOI] [PubMed] [Google Scholar]
  • 183.Thorp E, Cui D, Schrijvers DM, Kuriakose G, Tabas I. Mertk receptor mutation reduces efferocytosis efficiency and promotes apoptotic cell accumulation and plaque necrosis in atherosclerotic lesions of apoe−/− mice. Arterioscler Thromb Vasc Biol. 2008;28:1421–1428. doi: 10.1161/ATVBAHA.108.167197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Ait-Oufella H, Pouresmail V, Simon T, et al. Defective mer receptor tyrosine kinase signaling in bone marrow cells promotes apoptotic cell accumulation and accelerates atherosclerosis. Arterioscler Thromb Vasc Biol. 2008;28:1429–1431. doi: 10.1161/ATVBAHA.108.169078. [DOI] [PubMed] [Google Scholar]
  • 185.Canault M, Peiretti F, Kopp F, et al. The TNF alpha converting enzyme (TACE/ADAM17) is expressed in the atherosclerotic lesions of apolipoprotein E-deficient mice: possible contribution to elevated plasma levels of soluble TNF alpha receptors. Atherosclerosis. 2006;187:82–91. doi: 10.1016/j.atherosclerosis.2005.08.031. [DOI] [PubMed] [Google Scholar]
  • 186.Sather S, Kenyon KD, Lefkowitz JB, et al. A soluble form of the Mer receptor tyrosine kinase inhibits macrophage clearance of apoptotic cells and platelet aggregation. Blood. 2007;109:1026–1033. doi: 10.1182/blood-2006-05-021634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Cai B, Thorp EB, Doran AC, et al. MerTK cleavage limits proresolving mediator biosynthesis and exacerbates tissue inflammation. Proc Natl Acad Sci U S A. 2016;113:6526–6531. doi: 10.1073/pnas.1524292113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Cai B, Thorp EB, Doran AC, et al. MerTK receptor cleavage promotes plaque necrosis and defective resolution in atherosclerosis. J Clin Invest. 2017 doi: 10.1172/JCI90520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Garbin U, Baggio E, Stranieri C, et al. Expansion of necrotic core and shedding of Mertk receptor in human carotid plaques: a role for oxidized polyunsaturated fatty acids? Cardiovasc Res. 2013;97:125–133. doi: 10.1093/cvr/cvs301. [DOI] [PubMed] [Google Scholar]
  • 190.Rudensky AY. Regulatory T cells and Foxp3. Immunol Rev. 2011;241:260–268. doi: 10.1111/j.1600-065X.2011.01018.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Shevach EM. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. 2009;30:636–645. doi: 10.1016/j.immuni.2009.04.010. [DOI] [PubMed] [Google Scholar]
  • 192.Torchinsky MB, Garaude J, Martin AP, Blander JM. Innate immune recognition of infected apoptotic cells directs T(H)17 cell differentiation. Nature. 2009;458:78–82. doi: 10.1038/nature07781. [DOI] [PubMed] [Google Scholar]
  • 193.Sarang Z, Joos G, Garabuczi E, Ruhl R, Gregory CD, Szondy Z. Macrophages engulfing apoptotic cells produce nonclassical retinoids to enhance their phagocytic capacity. J Immunol. 2014;192:5730–5738. doi: 10.4049/jimmunol.1400284. [DOI] [PubMed] [Google Scholar]
  • 194.Chung EY, Liu J, Homma Y, et al. Interleukin-10 expression in macrophages during phagocytosis of apoptotic cells is mediated by homeodomain proteins Pbx1 and Prep-1. Immunity. 2007;27:952–964. doi: 10.1016/j.immuni.2007.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Lucas M, Stuart LM, Savill J, Lacy-Hulbert A. Apoptotic cells and innate immune stimuli combine to regulate macrophage cytokine secretion. J Immunol. 2003;171:2610–2615. doi: 10.4049/jimmunol.171.5.2610. [DOI] [PubMed] [Google Scholar]
  • 196.Lucas M, Stuart LM, Zhang A, et al. Requirements for apoptotic cell contact in regulation of macrophage responses. J Immunol. 2006;177:4047–4054. doi: 10.4049/jimmunol.177.6.4047. [DOI] [PubMed] [Google Scholar]
  • 197.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]
  • 198.Perruche S, Zhang P, Liu Y, Saas P, Bluestone JA, Chen W. CD3-specific antibody-induced immune tolerance involves transforming growth factor-beta from phagocytes digesting apoptotic T cells. Nat Med. 2008;14:528–535. doi: 10.1038/nm1749. [DOI] [PubMed] [Google Scholar]
  • 199.Mucida D, Pino-Lagos K, Kim G, et al. Retinoic acid can directly promote TGF-beta-mediated Foxp3(+) Treg cell conversion of naive T cells. Immunity. 2009;30:471–472. doi: 10.1016/j.immuni.2009.03.008. author reply 472–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Yamagiwa S, Gray JD, Hashimoto S, Horwitz DA. A role for TGF-beta in the generation and expansion of CD4+CD25+ regulatory T cells from human peripheral blood. J Immunol. 2001;166:7282–7289. doi: 10.4049/jimmunol.166.12.7282. [DOI] [PubMed] [Google Scholar]
  • 201.Fantini MC, Becker C, Monteleone G, Pallone F, Galle PR, Neurath MF. Cutting edge: TGF-beta induces a regulatory phenotype in CD4+CD25− T cells through Foxp3 induction and down-regulation of Smad7. J Immunol. 2004;172:5149–5153. doi: 10.4049/jimmunol.172.9.5149. [DOI] [PubMed] [Google Scholar]
  • 202.Chen W, Jin W, Hardegen N, et al. Conversion of peripheral CD4+CD25− naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003;198:1875–1886. doi: 10.1084/jem.20030152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 2007;204:1757–1764. doi: 10.1084/jem.20070590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Mucida D, Park Y, Kim G, et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007;317:256–260. doi: 10.1126/science.1145697. [DOI] [PubMed] [Google Scholar]
  • 205.Sun CM, Hall JA, Blank RB, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775–1785. doi: 10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol. 2012;30:531–564. doi: 10.1146/annurev.immunol.25.022106.141623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Tone Y, Furuuchi K, Kojima Y, Tykocinski ML, Greene MI, Tone M. Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer. Nat Immunol. 2008;9:194–202. doi: 10.1038/ni1549. [DOI] [PubMed] [Google Scholar]
  • 208.Zheng Y, Josefowicz S, Chaudhry A, Peng XP, Forbush K, Rudensky AY. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature. 2010;463:808–812. doi: 10.1038/nature08750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Jaensson E, Uronen-Hansson H, Pabst O, et al. Small intestinal CD103+ dendritic cells display unique functional properties that are conserved between mice and humans. J Exp Med. 2008;205:2139–2149. doi: 10.1084/jem.20080414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Johansson-Lindbom B, Svensson M, Pabst O, et al. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J Exp Med. 2005;202:1063–1073. doi: 10.1084/jem.20051100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol. 2007;8:1086–1094. doi: 10.1038/ni1511. [DOI] [PubMed] [Google Scholar]
  • 212.Murai M, Turovskaya O, Kim G, et al. Interleukin 10 acts on regulatory T cells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis. Nat Immunol. 2009;10:1178–1184. doi: 10.1038/ni.1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Paidassi H, Acharya M, Lacy-Hulbert A. Alpha (v) integrins license regulatory T cells to apoptotic cells and self-associated antigens. Ann N Y Acad Sci. 2010;1209:68–76. doi: 10.1111/j.1749-6632.2010.05783.x. [DOI] [PubMed] [Google Scholar]
  • 214.Munger JS, Huang X, Kawakatsu H, et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell. 1999;96:319–328. doi: 10.1016/s0092-8674(00)80545-0. [DOI] [PubMed] [Google Scholar]
  • 215.Paidassi H, Acharya M, Zhang A, et al. Preferential expression of integrin alphavbeta8 promotes generation of regulatory T cells by mouse CD103+ dendritic cells. Gastroenterology. 2011;141:1813–1820. doi: 10.1053/j.gastro.2011.06.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Worthington JJ, Czajkowska BI, Melton AC, Travis MA. Intestinal dendritic cells specialize to activate transforming growth factor-beta and induce Foxp3+ regulatory T cells via integrin alphavbeta8. Gastroenterology. 2011;141:1802–1812. doi: 10.1053/j.gastro.2011.06.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Boucard-Jourdin M, Kugler D, Endale Ahanda ML, et al. beta8 Integrin Expression and Activation of TGF-beta by Intestinal Dendritic Cells Are Determined by Both Tissue Microenvironment and Cell Lineage. J Immunol. 2016;197:1968–1978. doi: 10.4049/jimmunol.1600244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Travis MA, Reizis B, Melton AC, et al. Loss of integrin alpha(v)beta8 on dendritic cells causes autoimmunity and colitis in mice. Nature. 2007;449:361–365. doi: 10.1038/nature06110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Maeda A, Schwarz A, Kernebeck K, et al. Intravenous infusion of syngeneic apoptotic cells by photopheresis induces antigen-specific regulatory T cells. J Immunol. 2005;174:5968–5976. doi: 10.4049/jimmunol.174.10.5968. [DOI] [PubMed] [Google Scholar]
  • 220.Mougel F, Bonnefoy F, Kury-Paulin S, et al. Intravenous infusion of donor apoptotic leukocytes before transplantation delays allogeneic islet graft rejection through regulatory T cells. Diabetes Metab. 2012;38:531–537. doi: 10.1016/j.diabet.2012.08.008. [DOI] [PubMed] [Google Scholar]
  • 221.Huang FP, Platt N, Wykes M, et al. A discrete subpopulation of dendritic cells transports apoptotic intestinal epithelial cells to T cell areas of mesenteric lymph nodes. J Exp Med. 2000;191:435–444. doi: 10.1084/jem.191.3.435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Jang MH, Sougawa N, Tanaka T, et al. CCR7 is critically important for migration of dendritic cells in intestinal lamina propria to mesenteric lymph nodes. J Immunol. 2006;176:803–810. doi: 10.4049/jimmunol.176.2.803. [DOI] [PubMed] [Google Scholar]
  • 223.Tran DQ, Andersson J, Wang R, Ramsey H, Unutmaz D, Shevach EM. GARP (LRRC32) is essential for the surface expression of latent TGF-beta on platelets and activated FOXP3+ regulatory T cells. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:13445–13450. doi: 10.1073/pnas.0901944106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Guiducci C, Valzasina B, Dislich H, Colombo MP. CD40/CD40L interaction regulates CD4+CD25+ T reg homeostasis through dendritic cell-produced IL-2. Eur J Immunol. 2005;35:557–567. doi: 10.1002/eji.200425810. [DOI] [PubMed] [Google Scholar]
  • 225.Kumanogoh A, Wang X, Lee I, et al. Increased T cell autoreactivity in the absence of CD40-CD40 ligand interactions: a role of CD40 in regulatory T cell development. J Immunol. 2001;166:353–360. doi: 10.4049/jimmunol.166.1.353. [DOI] [PubMed] [Google Scholar]
  • 226.Weih F, Carrasco D, Bravo R. Constitutive and inducible Rel/NF-kappa B activities in mouse thymus and spleen. Oncogene. 1994;9:3289–3297. [PubMed] [Google Scholar]
  • 227.Azukizawa H, Dohler A, Kanazawa N, et al. Steady state migratory RelB+ langerin+ dermal dendritic cells mediate peripheral induction of antigen-specific CD4+ CD25+ Foxp3+ regulatory T cells. European journal of immunology. 2011;41:1420–1434. doi: 10.1002/eji.201040930. [DOI] [PubMed] [Google Scholar]
  • 228.Francisco LM, Salinas VH, Brown KE, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009;206:3015–3029. doi: 10.1084/jem.20090847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Guo Z, Jang MH, Otani K, et al. CD4+CD25+ regulatory T cells in the small intestinal lamina propria show an effector/memory phenotype. Int Immunol. 2008;20:307–315. doi: 10.1093/intimm/dxm143. [DOI] [PubMed] [Google Scholar]
  • 230.Raker VK, Domogalla MP, Steinbrink K. Tolerogenic Dendritic Cells for Regulatory T Cell Induction in Man. Front Immunol. 2015;6:569. doi: 10.3389/fimmu.2015.00569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Maldonado RA, von Andrian UH. How tolerogenic dendritic cells induce regulatory T cells. Adv Immunol. 2010;108:111–165. doi: 10.1016/B978-0-12-380995-7.00004-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Belladonna ML, Volpi C, Bianchi R, et al. Cutting edge: Autocrine TGF-beta sustains default tolerogenesis by IDO-competent dendritic cells. J Immunol. 2008;181:5194–5198. doi: 10.4049/jimmunol.181.8.5194. [DOI] [PubMed] [Google Scholar]
  • 233.Williams CA, Harry RA, McLeod JD. Apoptotic cells induce dendritic cell-mediated suppression via interferon-gamma-induced IDO. Immunology. 2008;124:89–101. doi: 10.1111/j.1365-2567.2007.02743.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Pallotta MT, Orabona C, Volpi C, et al. Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells. Nat Immunol. 2011;12:870–878. doi: 10.1038/ni.2077. [DOI] [PubMed] [Google Scholar]
  • 235.Ravishankar B, Liu H, Shinde R, et al. Tolerance to apoptotic cells is regulated by indoleamine 2,3-dioxygenase. Proc Natl Acad Sci U S A. 2012;109:3909–3914. doi: 10.1073/pnas.1117736109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Munn DH, Sharma MD, Baban B, et al. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity. 2005;22:633–642. doi: 10.1016/j.immuni.2005.03.013. [DOI] [PubMed] [Google Scholar]
  • 237.Baban B, Hansen AM, Chandler PR, et al. A minor population of splenic dendritic cells expressing CD19 mediates IDO-dependent T cell suppression via type I IFN signaling following B7 ligation. Int Immunol. 2005;17:909–919. doi: 10.1093/intimm/dxh271. [DOI] [PubMed] [Google Scholar]
  • 238.Baban B, Chandler PR, Sharma MD, et al. IDO activates regulatory T cells and blocks their conversion into Th17-like T cells. J Immunol. 2009;183:2475–2483. doi: 10.4049/jimmunol.0900986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Liu H, Huang L, Bradley J, et al. GCN2-dependent metabolic stress is essential for endotoxemic cytokine induction and pathology. Mol Cell Biol. 2014;34:428–438. doi: 10.1128/MCB.00946-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Ravishankar B, Liu H, Shinde R, et al. The amino acid sensor GCN2 inhibits inflammatory responses to apoptotic cells promoting tolerance and suppressing systemic autoimmunity. Proc Natl Acad Sci U S A. 2015;112:10774–10779. doi: 10.1073/pnas.1504276112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Nagata S. DNA degradation in development and programmed cell death. Annu Rev Immunol. 2005;23:853–875. doi: 10.1146/annurev.immunol.23.021704.115811. [DOI] [PubMed] [Google Scholar]
  • 242.Napirei M, Karsunky H, Zevnik B, Stephan H, Mannherz HG, Moroy T. Features of systemic lupus erythematosus in Dnase1-deficient mice. Nat Genet. 2000;25:177–181. doi: 10.1038/76032. [DOI] [PubMed] [Google Scholar]
  • 243.Chitrabamrung S, Rubin RL, Tan EM. Serum deoxyribonuclease I and clinical activity in systemic lupus erythematosus. Rheumatol Int. 1981;1:55–60. doi: 10.1007/BF00541153. [DOI] [PubMed] [Google Scholar]
  • 244.Yasutomo K, Horiuchi T, Kagami S, et al. Mutation of DNASE1 in people with systemic lupus erythematosus. Nat Genet. 2001;28:313–314. doi: 10.1038/91070. [DOI] [PubMed] [Google Scholar]
  • 245.Nagata S, Kawane K. Autoinflammation by endogenous DNA. Adv Immunol. 2011;110:139–161. doi: 10.1016/B978-0-12-387663-8.00004-1. [DOI] [PubMed] [Google Scholar]
  • 246.Sisirak V, Sally B, D'Agati V, et al. Digestion of Chromatin in Apoptotic Cell Microparticles Prevents Autoimmunity. Cell. 2016;166:88–101. doi: 10.1016/j.cell.2016.05.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Ullal AJ, Reich CF, 3rd, Clowse M, et al. Microparticles as antigenic targets of antibodies to DNA and nucleosomes in systemic lupus erythematosus. J Autoimmun. 2011;36:173–180. doi: 10.1016/j.jaut.2011.02.001. [DOI] [PubMed] [Google Scholar]
  • 248.Pisetsky DS, Lipsky PE. Microparticles as autoadjuvants in the pathogenesis of SLE. Nat Rev Rheumatol. 2010;6:368–372. doi: 10.1038/nrrheum.2010.66. [DOI] [PubMed] [Google Scholar]
  • 249.Wilber A, Lu M, Schneider MC. Deoxyribonuclease I-like III is an inducible macrophage barrier to liposomal transfection. Mol Ther. 2002;6:35–42. doi: 10.1006/mthe.2002.0625. [DOI] [PubMed] [Google Scholar]
  • 250.Ullal AJ, Marion TN, Pisetsky DS. The role of antigen specificity in the binding of murine monoclonal anti-DNA antibodies to microparticles from apoptotic cells. Clin Immunol. 2014;154:178–187. doi: 10.1016/j.clim.2014.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Barrat FJ, Meeker T, Gregorio J, et al. Nucleic acids of mammalian origin can act as endogenous ligands for Toll-like receptors and may promote systemic lupus erythematosus. J Exp Med. 2005;202:1131–1139. doi: 10.1084/jem.20050914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Lande R, Gregorio J, Facchinetti V, et al. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature. 2007;449:564–569. doi: 10.1038/nature06116. [DOI] [PubMed] [Google Scholar]
  • 253.Boule MW, Broughton C, Mackay F, Akira S, Marshak-Rothstein A, Rifkin IR. Toll-like receptor 9-dependent and -independent dendritic cell activation by chromatin-immunoglobulin G complexes. J Exp Med. 2004;199:1631–1640. doi: 10.1084/jem.20031942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Means TK, Latz E, Hayashi F, Murali MR, Golenbock DT, Luster AD. Human lupus autoantibody-DNA complexes activate DCs through cooperation of CD32 and TLR9. J Clin Invest. 2005;115:407–417. doi: 10.1172/JCI23025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Ganguly D, Chamilos G, Lande R, et al. Self-RNA-antimicrobial peptide complexes activate human dendritic cells through TLR7 and TLR8. J Exp Med. 2009;206:1983–1994. doi: 10.1084/jem.20090480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Leadbetter EA, Rifkin IR, Hohlbaum AM, Beaudette BC, Shlomchik MJ, Marshak-Rothstein A. Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors. Nature. 2002;416:603–607. doi: 10.1038/416603a. [DOI] [PubMed] [Google Scholar]
  • 257.Lau CM, Broughton C, Tabor AS, et al. RNA-associated autoantigens activate B cells by combined B cell antigen receptor/Toll-like receptor 7 engagement. J Exp Med. 2005;202:1171–1177. doi: 10.1084/jem.20050630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Marshak-Rothstein A, Rifkin IR. Immunologically active autoantigens: the role of toll-like receptors in the development of chronic inflammatory disease. Annu Rev Immunol. 2007;25:419–441. doi: 10.1146/annurev.immunol.22.012703.104514. [DOI] [PubMed] [Google Scholar]
  • 259.Kawane K, Fukuyama H, Kondoh G, et al. Requirement of DNase II for definitive erythropoiesis in the mouse fetal liver. Science. 2001;292:1546–1549. doi: 10.1126/science.292.5521.1546. [DOI] [PubMed] [Google Scholar]
  • 260.Kawane K, Ohtani M, Miwa K, et al. Chronic polyarthritis caused by mammalian DNA that escapes from degradation in macrophages. Nature. 2006;443:998–1002. doi: 10.1038/nature05245. [DOI] [PubMed] [Google Scholar]
  • 261.Yoshida H, Okabe Y, Kawane K, Fukuyama H, Nagata S. Lethal anemia caused by interferon-beta produced in mouse embryos carrying undigested DNA. Nat Immunol. 2005;6:49–56. doi: 10.1038/ni1146. [DOI] [PubMed] [Google Scholar]
  • 262.Baum R, Nundel K, Pawaria S, et al. Synergy between Hematopoietic and Radioresistant Stromal Cells Is Required for Autoimmune Manifestations of DNase II−/−IFNaR−/− Mice. J Immunol. 2016;196:1348–1354. doi: 10.4049/jimmunol.1502130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Okabe Y, Kawane K, Akira S, Taniguchi T, Nagata S. Toll-like receptor-independent gene induction program activated by mammalian DNA escaped from apoptotic DNA degradation. J Exp Med. 2005;202:1333–1339. doi: 10.1084/jem.20051654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Motani K, Ito S, Nagata S. DNA-Mediated Cyclic GMP-AMP Synthase-Dependent and -Independent Regulation of Innate Immune Responses. J Immunol. 2015;194:4914–4923. doi: 10.4049/jimmunol.1402705. [DOI] [PubMed] [Google Scholar]
  • 265.Ahn J, Gutman D, Saijo S, Barber GN. STING manifests self DNA-dependent inflammatory disease. Proc Natl Acad Sci U S A. 2012;109:19386–19391. doi: 10.1073/pnas.1215006109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Chan MP, Onji M, Fukui R, et al. DNase II-dependent DNA digestion is required for DNA sensing by TLR9. Nat Commun. 2015;6:5853. doi: 10.1038/ncomms6853. [DOI] [PubMed] [Google Scholar]
  • 267.White MJ, McArthur K, Metcalf D, et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell. 2014;159:1549–1562. doi: 10.1016/j.cell.2014.11.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Rongvaux A, Jackson R, Harman CC, et al. Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA. Cell. 2014;159:1563–1577. doi: 10.1016/j.cell.2014.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Baum R, Sharma S, Carpenter S, et al. Cutting edge: AIM2 and endosomal TLRs differentially regulate arthritis and autoantibody production in DNase II-deficient mice. J Immunol. 2015;194:873–877. doi: 10.4049/jimmunol.1402573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Pawaria S, Moody K, Busto P, et al. Cutting Edge: DNase II deficiency prevents activation of autoreactive B cells by double-stranded DNA endogenous ligands. J Immunol. 2015;194:1403–1407. doi: 10.4049/jimmunol.1402893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Pawaria S, Sharma S, Baum R, et al. Taking the STING out of TLR-driven autoimmune diseases: good, bad, or indifferent? J Leukoc Biol. 2017;101:121–126. doi: 10.1189/jlb.3MR0316-115R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Sharma S, Campbell AM, Chan J, et al. Suppression of systemic autoimmunity by the innate immune adaptor STING. Proc Natl Acad Sci U S A. 2015;112:E710–717. doi: 10.1073/pnas.1420217112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Huang L, Li L, Lemos H, et al. Cutting edge: DNA sensing via the STING adaptor in myeloid dendritic cells induces potent tolerogenic responses. J Immunol. 2013;191:3509–3513. doi: 10.4049/jimmunol.1301419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Lemos H, Huang L, McGaha TL, Mellor AL. Cytosolic DNA sensing via the stimulator of interferon genes adaptor: Yin and Yang of immune responses to DNA. Eur J Immunol. 2014;44:2847–2853. doi: 10.1002/eji.201344407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Pollard JW. Trophic macrophages in development and disease. Nat Rev Immunol. 2009;9:259–270. doi: 10.1038/nri2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496:445–455. doi: 10.1038/nature12034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Okabe Y, Medzhitov R. Tissue biology perspective on macrophages. Nat Immunol. 2016;17:9–17. doi: 10.1038/ni.3320. [DOI] [PubMed] [Google Scholar]
  • 278.Wood W, Martin P. Macrophage Functions in Tissue Patterning and Disease: New Insights from the Fly. Dev Cell. 2017;40:221–233. doi: 10.1016/j.devcel.2017.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Hanayama R, Nagata S. Impaired involution of mammary glands in the absence of milk fat globule EGF factor 8. Proc Natl Acad Sci U S A. 2005;102:16886–16891. doi: 10.1073/pnas.0508599102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Lee CS, Penberthy KK, Wheeler KM, et al. Boosting Apoptotic Cell Clearance by Colonic Epithelial Cells Attenuates Inflammation In Vivo. Immunity. 2016;44:807–820. doi: 10.1016/j.immuni.2016.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Monks J, Rosner D, Geske FJ, et al. Epithelial cells as phagocytes: apoptotic epithelial cells are engulfed by mammary alveolar epithelial cells and repress inflammatory mediator release. Cell Death Differ. 2005;12:107–114. doi: 10.1038/sj.cdd.4401517. [DOI] [PubMed] [Google Scholar]
  • 282.Monks J, Smith-Steinhart C, Kruk ER, Fadok VA, Henson PM. Epithelial cells remove apoptotic epithelial cells during post-lactation involution of the mouse mammary gland. Biol Reprod. 2008;78:586–594. doi: 10.1095/biolreprod.107.065045. [DOI] [PubMed] [Google Scholar]
  • 283.Poon IK, Lucas CD, Rossi AG, Ravichandran KS. Apoptotic cell clearance: basic biology and therapeutic potential. Nat Rev Immunol. 2014;14:166–180. doi: 10.1038/nri3607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Han CZ, Juncadella IJ, Kinchen JM, et al. Macrophages redirect phagocytosis by non-professional phagocytes and influence inflammation. Nature. 2016;539:570–574. doi: 10.1038/nature20141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Bourdonnay E, Zaslona Z, Penke LR, et al. Transcellular delivery of vesicular SOCS proteins from macrophages to epithelial cells blunts inflammatory signaling. J Exp Med. 2015;212:729–742. doi: 10.1084/jem.20141675. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES