Abstract
In recent years, researchers have devoted much attention to the diverse roles of macrophages and their contributions to tissue development, wound healing, and angiogenesis. What should not be lost in the discussions regarding the diverse biology of these cells is that when perturbed, macrophages are the primary contributors to potentially pathological inflammatory processes. Macrophages stand poised to rapidly produce large amounts of inflammatory cytokines in response to danger signals. The production of these cytokines can initiate a cascade of inflammatory mediator release that can lead to wholesale tissue destruction. The destructive inflammatory capability of macrophages is amplified by exposure to exogenous interferon-γ, which prolongs and heightens inflammatory responses. In simple terms, macrophages can thus be viewed as incendiary devices with hair triggers waiting to detonate. We have begun to ask questions about how these cells can be regulated to mitigate the collateral destruction associated with macrophage activation.
Keywords: cytokines, transcription, prostaglandin, adenosine, TLR, NF-κB
MACROPHAGES CONTRIBUTE TO INFLAMMATION
Macrophages are major producers of inflammatory mediators during autoimmune and autoinflammatory diseases. The kinase-dependent signaling pathways that emanate from ligated pattern recognition receptors on macrophages are well described. These pathways activate latent transcription factors, including NF-κB, CREB, AP-1, and interferon-regulatory factors (IRFs), to initiate cytokine gene transcription. However, recent studies suggest that the pattern of cytokine production by macrophages in response to inflammatory stimuli is more complex than simply the result of the activation of transcription factors (1). Macrophages are poised to rapidly respond to pathogen-associated molecular patterns (PAMPs) with the production of tumor necrosis factor (TNF) because RNA polymerase II is already positioned on the TNF gene in the basal state, capable of initiating transcription (2). Furthermore, the TNF promoter has relatively low histone occupancy, and the histones that are associated with the TNF promoter contain the activation marks H3K4me and H3K9Ac (3). Thus, signal-dependent transcription factor activation following receptor ligation, in concert with the positioning and covalent modification of nucleosomes adjacent to the TNF promoter, contributes to the rapid stimulus-dependent transcription of this cytokine gene.
In macrophages, the production of TNF usually precedes and generally promotes the carefully orchestrated release of many other inflammatory mediators, including interleukin (IL)-6, IL-12/23(p40), and the type I interferons. Chemokines, lipid mediators, and antimicrobial peptides are similarly released from stimulated macrophages. Many cells can change their primary transcriptional program in response to danger signals, such as lipopolysaccharide (LPS), but it is the production of these subsequent inflammatory mediators downstream from TNF that makes the macrophage such an important contributor to the inflammatory response. Macrophages are uniquely adept at producing a cascade of inflammatory cytokines and mediators in response to LPS exposure. This is largely due to the presence of lineage-specific transcription factors, such as PU.1 and C/EBP, which decorate enhancer regions upstream from cytokine genes and are thought to hold chromatin in an open configuration, allowing stimulus-dependent transcription factors to bind and promote gene expression (4). Thus, lineage-specific enhancers upstream from many inflammatory genes confer macrophage-specific responsiveness to inflammatory stimuli, and they define the anatomical specificity of macrophage responses in different tissues (5). The second wave of genes produced in response to stimulation usually also depends on enhancer activation, chromatin remodeling, and nucleosome repositioning to allow new transcription factor access. These processes can take time; therefore, one typically sees distinct temporal waves of gene expression following stimulation, sometimes designated as primary and secondary genes (2). Thus, cooperation between lineage-specific and stimulus-activated transcription factors, along with the repositioning of nucleosomes and covalent modifications to histones, determines how a given macrophage population will respond to inflammatory stimuli. Consequently, the chromatin in macrophages residing in the liver and the lung will be differentially “marked,” and therefore these two cells will not produce identical responses to a given stimuli.
INTERFERON-γ, A MAJOR MEDIATOR OF MACROPHAGE INFLAMMATION
Virtually all of these inflammatory responses are accentuated by the presence of interferon-γ (IFN-γ), which dramatically increases the production of inflammatory mediators by macrophages. This increase in cytokine production by IFN-γ-primed macrophages is due primarily to the activation of STAT1. The binding of interferons to their cognate receptors triggers tyrosine phosphorylation of STAT1, its localization to the nucleus, and DNA binding, leading to the transcription of at least 200 genes (6). Among these are mostly inflammation-associated genes, such as GM-CSF, IL-12p40, TNF-α, and IL-6. STAT1 binding increases macrophage inflammatory responses by many diverse mechanisms. It induces covalent modifications to histones to assume an active conformation, allowing the assembly of transcription complexes on inflammatory cytokine genes (7). IFN-γ priming can synergize with Toll-like receptor (TLR) signaling to enhance NF-κB-dependent gene expression and increase the stability of mRNAs encoding inflammatory mediators (8). Recent work indicates that IFN-γ treatment of macrophages also results in a more efficient translation of inflammatory mediators (9). Finally, IFN-γ-mediated STAT1 signaling potentiates inflammatory signaling by inhibiting IL-10 secretion. IFN-γ suppresses many of the key molecules required for IL-10 secretion, such as SOCS3, the TLR2-induced activation of the PI3K-Akt pathway (10), and the transcription factors CREB and AP-1 through a glycogen synthase kinase 3 (GSK3)-dependent mechanism (11).
IFN/STAT1 signaling can enhance the pathogenesis of septic shock (12), mediate pouchitis in inflammatory bowel diseases (13), and increase auto-antibody production in systemic lupus erythematosus (14). Recently, dominant gain of function STAT1 (GOF-S1) mutations in humans, characterized by the hyperphosphorylation of STAT1, were associated with an autoimmune phenotype with IPEX-like syndrome and a heightened susceptibility to fungal pathogens (15).
The increased production of TNF-α, IL-6, and IL-12 by macrophages following IFN-γ exposure is associated with an enhanced capacity for macrophages to kill intracellular pathogens and the development of long-lasting cellular immunity. IFN-γ is thus a hallmark cytokine of Th1 immune responses, and individuals with deficiencies in IFN-γ signaling are susceptible to a variety of intracellular infections (16). However, macrophages with interferon or STAT1 signatures were observed in a number of autoimmune diseases where they are the primary contributors to immunopathology. In rheumatoid arthritis, synovial macrophages express numerous IFN-γ inducible genes (17) and exhibit evidence of STAT1 activation. Increased levels of IFN-γ were also detected in the skin of lupus patients (18). Similarly, IFN-γ production by Th1 cells contributes to neurodegeneration in a murine model of Alzheimer’s disease (19). In psoriasis, a Th17 disease, serum IFN-γ levels are elevated and correlate with disease severity (20). In multiple sclerosis (MS), another Th17 disease, the role of classically activated macrophages and IFN-γ production appears to be somewhat more complex and can vary depending on the disease phase (21). Early studies identified IFN-γ-producing Th17 cells in lesions (22), and in progressive MS, patients typically have higher serum levels of IFN-γ and TNF (23). However, in the murine model of MS, called experimental autoimmune encephalomyelitis (EAE), mice lacking IFN-γ are more susceptible to severe EAE, and the administration of antibodies to IFN-γ actually exacerbates disease (21). One explanation for this is that IFN-γ production by astrocytes rather than microglia may preserve neuronal integrity (24), but the other obvious explanation is that IFN-γ may be protective by virtue of its well-described inhibition of (pathological) Th17 responses. Regardless of the mechanism of induction, activated macrophages are key contributors to autoimmune pathology in all of the aforementioned diseases.
Fortunately, several types of molecular controls work to downregulate the inflammatory responses of activated macrophages. These include inhibitors of signaling components associated with pattern recognition receptors, the activation of transcription factors that can repress gene expression, epigenetic silencing of gene expression, and the production of miRNAs to diminish inflammatory responses. These regulatory controls are the focus of this review.
NEGATIVE REGULATION OF TOLL-LIKE RECEPTOR SIGNALING
A variety of regulatory mechanisms have evolved to protect the immune system from aberrant TLR signaling. Most of these inhibitory proteins are inducible, suggesting a negative feedback on TLR signaling pathways. Some of these known negative regulators with their mechanism of action and their relevance to disease are listed in Table 1. The negative regulation of TLR responses is particularly important in the gut where it prevents unwanted immune responses to the normal flora. Because sustained signaling of TLRs can result in autoimmune and chronic inflammatory diseases, the tight regulation of the TLR signaling pathway is crucial not only to suspend the pathway when danger is removed but also to maintain homeostasis in those anatomical areas of constant exposure to normal flora. Negative regulation of TLR signaling can happen at any step in TLR signaling, from surface sensing of ligands to the transcription of the downstream effector molecules.
Table 1.
Negative regulators of Toll-like receptor (TLR) signaling with their mechanism of action and relevance to disease
Negative regulators | Mode of action | Diseases implicated | Reference(s) |
---|---|---|---|
Regulators of ligand binding | |||
sTLR2 (soluble form of TLR2) | Prevents lipopeptide binding to TLR2 through competitive binding | In human milk sTLR2 protects infants from enterocolitis and sepsis | 139 |
sTLR4 (soluble form of TLR4) | Prevents LPS binding to TLR4 through competitive binding | Elevated levels of sTLR4 are found in human endotoxemia and auto-inflammatory syndromes | 140 |
sCD14 (soluble CD14) also known as presepsin | Diverts LPS from membrane-bound CD14 and promotes LPS efflux from cell-surface CD14 | Marker of inflammatory diseases and sepsis | 141, 142 |
RP105 (radioprotective 105) | RP105-MD1 complex binds to TLR4-MD2 complex and prevents LPS binding | Increased expression in primary biliary cirrhosis | 25, 143 |
Triad3A | E3 ubiquitin-protein ligase that enhances ubiquitination and degradation of TLR4 and TLR9 | Overexpression reported in diabetic kidneys | 144, 145 |
Regulators of adaptor complexes | |||
SARM (sterile alpha and armadillo motif-containing molecule) | Negatively regulates TRIF-dependent TLR signaling and MyD88-mediated AP-1 activation | May mediate endotoxin tolerance | 146 |
Splicing variant of MyD88 (MyD88s) | Splice form is unable to activate NF-κB | None | 27 |
TAG | Splice variant of TRAM, negatively regulates MyD88-independent TLR4 pathway | None | 28 |
ST2 (single immunoglobulin IL-1R-related molecule) ST2L: membrane bound ST2: soluble form |
Prevents the recruitment of MyD88 and TIRAP (MAL) to TLR2, -4, and -6 by sequestering them | Prevents renal epithelial immunity in diabetes | 145, 147 |
TRAF1 (TNF receptor-associated factor) | Inhibits TLR3 mediated NF-κB activation and IFN-β signaling by interacting with TRIF | TRAF1 polymorphisms are associated with autoimmune thyroid disease | 148, 149 |
TRAF4 | Physically interacts and functionally counteracts with TRAF6 and TRIF | Implicated in human IBD | 150, 151 |
Cbl-b (casitas B-lineage lymphoma, an E3 ubiquitin ligase) | Mediates ubiquitination of MyD88 and TRIF | Causes dysregulation of macrophage activation in obesity-induced insulin resistance | 152, 153 |
CYLD (deubiquitinase) | By deubiquitinating K63-linked polyubiquitination of MyD88, a form required for TLR signaling | Brooke-Spiegler syndrome | 154, 155 |
SOCS1 | Phosphorylated TIRAP (MAL) is bound to SOCS1 which results in TIRAP polyubiquitination and degradation | Psoriasis, MS, viral infections, and many others | 156 |
Regulators of signaling molecules | |||
Splicing variants of IRAK1 (IRAK1c), IRAK2 (IRAK2c, 2d) | Splice forms were unable to activate NF-κB | None | 157, 158 |
IRAKM | Lacks intrinsic kinase activity and hence prevents dissociation of IRAK4 and IRAK1 from MyD88 | IRAKM+22148 G>A allele associated with risk of developing chronic relapsing pouchitis | 159, 160 |
SHP-1 and SHP-2 (Src homology 2 domain-containing protein tyrosine phosphatase-1 and -2) | SHP-1 suppresses IRAK1 and IRAK2 activities and SHP-2 negatively regulates TRIF-dependent type I IFN production | Leishmania spp. promote SHP-1 binding to IRAK1 to suppress innate response | 161, 162 |
SIGIRR (single immunoglobulin interleukin-1 receptor-related) | Interacts with and blocks the activation of IRAKs and TRAF6 | SLE, dominant negative forms reported in human colon tumors | 163 |
A20 (deubiquitinase) | Deubiquitinates TRAF6 and prevents its binding to TAK1 and subsequent Iκκ complex formation | TNFAIP3 gene (that codes for A20) polymorphisms are reported in RA, SLE, psoriasis, Crohn’s disease, and other autoinflammatory diseases | 30, 164 |
β-Arrestin 2 | Interacts with TRAF6 and prevents its oligomerizationNegatively regulates TLR4 signaling by targeting p38 MAPK and IL-10; regulates TLR4-mediated apoptotic signaling through GSK3α | Several fibrotic diseases | 165, 166 |
TRIM38 (tripartite-motif containing 38) | Binds to TRAF6 and promotes K48-linked polyubiquitination, and proteasomal degradation | Primary Sjögren’s syndrome | 167, 168 |
Lyp (lymphocyte tyrosine phosphatase) | Potentiates type I IFN from TLR signaling through interaction with TRAF3 and promoting its K63 ubiquitination | SNP in PTPN22 gene that encodes Lyp results in autoimmune risk allele (Lyp620W) and hence associated with several autoimmune diseases | 169 |
TRIM30α (tripartite-motif containing −30α) | Negatively regulates NF-κB activation by targeting TAB2 and TAB3 for degradation | DSS-induced colitis mouse model | 170 |
Regulators of transcription factors | |||
ATF3 (activating transcription factor 3; a member of ATF/DREB family) | Restrict access to NF-κB and AP-1 to the promoter regions of proinflammatory cytokines IL-6 and IL-12b | Type 2 diabetes, cardiac hypertrophy, osteoarthritis | 35, 171 |
NOD2 | NOD2 signaling inhibits TLR2 driven activation of NF-κB subunit c-Rel | Crohn’s disease, sarcoidosis | 172, 173 |
PI3K | Suppresses p38 MAPK and NF-κB through PKB; PI3K can also directly inhibit IL-12 production | E.g., fibrosis, diabetes, COPD, IBD, RA, SLE, MS | 10, 174 |
Pin1 (peptidyl prolyl isomerase) | Mediates ubiquitination and subsequent proteasome-dependent degradation of IRF3 | Overexpressed in several cancers; implicated in allergic pulmonary inflammation | 175, 176 |
IRF4 | Competitively inhibits IRF5 binding to TLR | SLE | 177, 178 |
RAUL (RTA-associated ubiquitin ligase) | Catalyzes ubiquitination of IRF3/7 and negatively regulates type I IFN responses | Kaposi sarcoma herpes virus stabilizes RAUL by preventing its degradation and thereby downmodulating antiviral response | 179 |
Abbreviations: COPD, chronic obstructive pulmonary disease; DSS, dextran sulfate sodium; IBD, inflammatory bowel disease; IFN, interferon; IRF, interferon regulatory factor; LPS, lipopolysaccharide; MAL, MyD88 adapter-like; MS, multiple sclerosis; PKB, protein kinase B; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus; SNP, single nucleotide polymorphism; TAG, TRAM adaptor with GOLD domain; TIRAP, Toll-like receptor adaptor protein.
Regulators of Ligand Binding
Members of this class either prevent the binding of ligand to membrane TLRs or displace them when bound. This family includes the soluble TLR receptors (sTLRs) sTLR2 and sTLR4 that act as decoy receptors, sCD14 that binds directly to LPS, and radioprotective 105 (RP105), a TLR homolog that prevents TLR4 from binding to LPS. All are negative regulators of TLR signaling. The binding of soluble forms of TLRs to their free ligands in plasma prevents ligand binding to surface TLRs. The high levels of sTLRs in human breast milk to prevent colitis in infants represent one of the best examples of this type of inhibition. The soluble form of coreceptor sCD14 binds to and diverts LPS from membrane-bound CD14. sCD14 also promotes LPS efflux from cell-surface CD14, transferring it to plasma lipoproteins. The role of RP105 may be cell type specific, but in myeloid cells, RP105/MD-1 interacts directly with TLR-4/MD-2 to inhibit the ability of this signaling complex to bind LPS (25). Triad3A, a RING finger protein, acts as an E3 ubiquitin-protein ligase and enhances ubiquitination and proteolytic degradation of TLR4 and TLR9. Thus, it renders receptors inaccessible to their ligands (Figure 1).
Figure 1.
Inflammation can be negatively regulated by a multitude of cellular molecules and mechanisms. The initiation of TLR signaling pathways leading to inflammation can be negatively regulated at various levels from ligand binding to transcription. Whereas primary or secondary signaling through certain GPCRs, nuclear receptors, or inhibitory receptors can cross-regulate the inflammatory pathways, cellular mechanisms such as epigenetic regulation, ubiquitination, and cell death can operate at both micro- and macrolevels to inhibit inflammation in macrophages. Such receptor-mediated signaling pathways and cellular mechanisms can give rise to anti-inflammatory cytokines that can mitigate proinflammatory cytokines; transcription factors that turn on mRNA transcription of anti-inflammatory molecules; and microRNAs that mediate post-transcriptional silencing of mRNAs encoding inflammatory proteins. Abbreviations: CYLD, cylindromatosis; ER, endoplasmic reticulum; FcγR, Fcγ receptor; GPCR, G-protein coupled receptor; IFN-α, interferon-α; IL, interleukin; LYP, lymphocyte tyrosine phosphatase; PPARγ, peroxisome proliferator-activated receptor γ; TGF-β, transforming growth factor-β; TLR, Toll-like receptor.
Regulators of Adaptor Complexes
TLR signaling includes five adaptors known as MyD88, MAL (TIRAP), TRIF, TRAM, and SARM, all of which contain a Toll/Il-1 receptor (TIR) domain (26). Both MyD88 and TRIF bind directly to TLRs, whereas MAL (adaptor-like MyD88) and TRAM act as recruiting adaptors to bring MyD88 and TRIF to TLRs, respectively. MyD88-mediated signaling results in NF-κB activation, whereas TRIF-mediated signaling results in IRF activation. More inhibitory mechanisms exist for the MAL-MyD88 adaptor complex compared to the TRIF-TRAM complex, as the former is utilized by almost all TLRs (except TLR3), and the latter complex is utilized only by TLR3 and TLR4. Inhibitors to these adaptors include (a) negatively regulating adaptors, (b) variants of the adaptor proteins, (c) proteins that directly bind to and prevent adaptor recruitment or functions, and (d) proteins that target adaptor proteins for degradation. SARM is the only negative TIR domain containing an adaptor that negatively regulates TRIF-dependent TLR signaling and MyD88-mediated AP-1 activation. The presence of this adaptor is implicated in endotoxin tolerance. The splice variants of MyD88, termed MyD88s, and of TRAM, known as TAG (TRAM adaptor with GOLD domain), are two variants of the adaptor complexes that negatively regulate the two signaling arms of TLR (27, 28). Proteins such as ST2L, TRAF1, and TRAF4 directly bind to the adaptor molecules and negatively regulate their functions. Whereas ST2 prevents the recruitment of MyD88 to TIRAP, TRAFs prevent the association of TRIFs to TLRs. Cbl-b ubiquitin ligase targets adaptors of both pathways via MyD88 and TRIF for ubiquitination, thereby negatively regulating them. Although SOCS1 does not directly mediate ubiquitination, binding of MAL to SOCS1 triggers its polyubiquitination and degradation.
Regulators of Signaling Molecules
This class of molecules inhibits TLR signaling pathway components that lie between the adaptors and the transcription factors. These include IRAKs, TRAF3, TRAF6, TBK1, and TAB2/3. Of these molecules, TRAF6 is of primary importance, as almost all TLR pathways converge at TRAF6 for subsequent signaling. Most of the negative regulation at this level occurs by targeting TRAF6 for ubiquitination/deubiquitination, resulting in varying mechanisms of ubiquitination-dependent inhibition. Interaction of TRAF6 with β-arrestin 2 prevents its oligomerization, autoubiquitination, and subsequent activation of NF-κB and AP-1 (29). In contrast, deubiquitination of TRAF6 by A20 prevents its binding to TAK1 and subsequent formation of the Iκκ complex (30). TRIM38, a tripartite motif-containing molecule, promotes TRAF6 ubiquitination by directly binding to it and targeting it for proteasomal degradation. The observations that multiple negative regulators exist for TRAF6 indicate the essential and central role of this protein in TLR signaling. The inhibition of IRAK kinases can occur through the competitive binding of the splice variants IRAK1c, IRAK2c, IRAK2d, and IRAKM, which competitively bind to adaptor molecules and inhibit IRAK signaling. The Src homology 2 domain-containing proteins tyrosine phosphatase-1 (SHP-1) and phosphatase-2 (SHP-2) suppress the IRAK1 and IRAK2 activity.
Regulators of Transcription
Sensing of PAMPs and danger-associated molecular patterns by the TLR family leads to a cascade of events, most of which converge at transcription factors NF-κB and IRF3/7. The NF-κB family of transcription factors is central to the macrophage inflammatory response. This family of transcription factors is a target for regulation owing to the large number of genes it transcribes. The negative regulation strategies include preventing the binding of NF-κB to target genes, regulating the assembly of its members and inhibiting their transcription, and averting post-translational modifications and protein degradation (31).
B cell lymphoma 6 (Bcl-6) is an important negative regulator of NF-κB signaling. Nearly one-third of the LPS-induced genes are regulated by Bcl-6 (32). This transcriptional repressor also binds to IRF7 loci to restrain antiviral responses. The transcription of key inflammatory chemokine genes in macrophages is also inhibited by Bcl-6 (33). Consequently, Bcl-6-null mice have a lethal inflammatory phenotype. The activation of peroxisome proliferator-activated receptor delta (PPARδ) results in increased macrophage Bcl-6 levels to decrease their inflammatory potential.
In LPS signaling, SOCS1 was shown to decrease NF-κB signaling through the ubiquitination complex known as ECS-SOCS1. This complex, which is triggered by COMMD1, is composed of multimeric ubiquitin ligases, containing elongins B and C, Cul2, and SOCS. Through this complex, COMMD1 accelerates the ubiquitination and degradation of NF-κB subunits (34). Activating transcription factor 3 (ATF3) is a type I IFN-inducible protein that recruits histone deacetylases to alter chromatin structure and restrict access to NF-κB and AP-1 to the promoter regions of proinflammatory cytokines (35).
Inhibition of IRF functions in TLR signaling can occur through ubiquitination-dependent IRF degradation or via negatively regulating IRFs. Stimulation of TLRs with dsRNA can lead to Ser339-Pro340 phosphorylation of IRF3. Peptidyl prolyl isomerase 1 interacts with this phosphorylated form of IRF3 and polyubiquitinates the transcription factor, targeting it for proteasome degradation. Negatively regulating IRFs include IRF4 that competitively inhibits IRF5 from binding to TLR, thereby inhibiting inflammatory responses.
OTHER TRANSCRIPTIONAL REGULATORS
Among the transcription factors that are associated with TLR signaling, the STATs play a particularly important role. Whereas STAT1, STAT2, STAT4, and STAT5 signal and mediate proinflammatory responses, STAT3 and STAT6 transcribe anti-inflammatory genes. The best studied is STAT3, which is a key transcriptional regulator of IL-10, a major anti-inflammatory cytokine. This transcription factor can uniquely induce both anti-inflammatory and proinflammatory responses within the same cell. When signaled via IL-10, it induces anti-inflammatory responses, and when stimulated by IL-6, it drives inflammation. In macrophages, STAT3 combines with a specific set of transcription factors such as PU.1, CEBPA, and CEBPB, which are prebound to sites of future STAT3 recruitment (36), thus mediating anti-inflammation. The reciprocal cross talk between STAT3 and the p38 mitogen-activated protein kinase (MAPK) pathway is critical in macrophages to balance the induction, propagation, and resolution of LPS signaling. The IL-10 production that occurs later during TLR signaling triggers STAT3 phosphorylation and activation. STAT3 then drives the expression of DUSP1, which is a negative regulator of the p38 MAPK, thus closing the feedback loop (37). The importance of STAT3 in anti-inflammation is evident from the clinical manifestations associated with STAT3 mutations in humans. Loss-of-function STAT3 in humans results in hyper–immunoglobulin E (IgE) syndrome, which is characterized by both primary immunodeficiency and a severe autoimmune phenotype with high proinflammatory gene transcripts (38).
The binding of cytokines IL-4 and IL-13 to their receptors triggers STAT6 phosphorylation, leading to the alternate activation of macrophages. Phosphorylated STAT6 translocates to the nucleus and transcribes several genes required for alternate activation of macrophages, such as resistin-like α (Retnlα, Fizz1) and chitinase 3-like 3 (Chi3l3, Ym1), peroxisome proliferator-activated receptor γ (PPARγ), mannose receptor (Mrc1) (39), and genes that are involved in β oxidation of fatty acids. STAT6 also synergizes with Krüppel-like factor 4 and PPARγ to inhibit the inflammatory genes TNF-α, Cox-2, CCL5, and iNOS (40). In AA-Mφ, IL-4 induces the immune inhibitory ligand and the programmed death ligand 2 via STAT6, the surface expression of which leads to the inhibition of Th2 proliferation (41). Numerous studies have demonstrated the important protective role of AA-Mφ and thus STAT6 signaling in host immunity to helminthic infection and in wound healing (42).
microRNA REGULATION OF MACROPHAGES IN INFLAMMATION AND AUTOIMMUNITY
The levels of several TLR signaling molecules are regulated at the transcriptional level by noncoding RNAs, such as microRNA (miRNA). miRNAs are small, approximately 20–24-nucleotide-long RNAs that generally bind to 3 untranslated regions of mRNAs. They have emerged as critical regulators of macrophage functions and polarization. Studies have profiled miRNA expression in polarized macrophages to better understand their biological function. In general, miRNA profiling shows significant differences between human and mouse macrophages. For example, stimulation of murine bone marrow derived macrophages with IFN-γ+LPS upregulates miR-127-3p, miR-155-5p, miR-181a, miR-204-5p, and miR-451, whereas in human macrophages, miR-27a, miR-29b, miR-125a, miR-146a, and miR-155 are upregulated. Similarly, AA-Mϕ of mouse upregulate miR-125-5p, miR-143-3p, miR-145-5p, and miR-146a-3p, whereas human AA-Mϕ upregulate a different set of miRNAs, including miR-26a and miR-193b (43, 44).
Classification of miRNAs as pro- and anti-inflammatory is difficult owing to wide heterogeneity in miRNA functions even among members of the same family. Whereas some miRNAs, such as miR155, have a clear-cut proinflammatory role, others have dual roles depending on the cell type, stimulus, and experimental conditions. Nevertheless, considerable evidence demonstrates the important roles for some of these molecules in regulating macrophage inflammatory activity.
miR-124 is associated with decreased macrophage inflammation. The anti-inflammatory role of this miRNA was first demonstrated in microglial cells during EAE (45). In this model, the overexpression of miR-124 led to direct inhibition of the transcription factor CCAAT/enhancer-binding protein-α and its downstream target PU.1, resulting in the deactivation of microglial cells. Furthermore, treatment with miR-124 reduced the severity of EAE and the percentage of activated macrophages. The same group studied the role of miR-124 in allergy and asthma, demonstrating that IL-4/IL-13 induced the expression of miR-124 in human macrophages, which led to increased allergic inflammation. In human patients with allergies and bronchial asthma, CD14+CD16+ intermediate monocytes expressing miR-124 increased in number (46). The miRNA-146 family of miRNAs, consisting of miRNA-146a and miRNA-146b, consists of negative feedback regulators of TLR signaling. miRNA-146a targets IRAK1, TRAF6, IRAK2, and IRF5, and miRNA-146b targets TLR4, MyD88, IRAK1, and TRAF6 (47). miRNA-146b is an IL-10-dependent miRNA that regulates the LPS-mediated proinflammatory cytokine secretion of TNF-α, IL-6, and IL-8 and chemokines CCL2, CCL3, and IP-10 (48). miRNA-147 regulates inflammatory responses in murine macrophages, and in breast cancer, it suppresses the AKT/mTOR signaling pathway (49). Its relevance in human inflammatory diseases has yet to be uncovered. miR-187 is directly induced by IL-10 signaling. It recruits TNFA mRNA to the RISC complex to promote its degradation. It also negatively regulates IL-6 and IL-12p40 production in TLR4-stimulated monocytes (50). Due to its ability to favor an M2 phenotype, miR-187 is overexpressed in long-term asthma and allergic rhinitis in humans (51). The miR-373-3P is located in an intronic region of PPARγ-coactivator-1β, a protein involved in the alternative activation of macrophages. Thus, this miRNA is conceivably involved in the alternate activation of macrophages (52).
THE REGULATION OF INFLAMMATION BY ENDOGENOUS REGULATORY MOLECULES
The wave of inflammatory mediators released by macrophages in response to pathogen products is an important innate response to invading pathogens. However, the return of macrophages to homeostasis is equally as important, because when left unchecked, macrophage activation can lead to destructive tissue pathology. In the late stages of some infections, macrophages can be exposed to molecules that not only mitigate inflammation but actively return the macrophage to a resting state. Many of these molecules are produced by macrophages themselves, and we propose that the synthesis and response to these endogenous mediators are what typically induce a transient state of macrophage activation. This transient activation is a way that macrophages can respond to danger and yet still contribute to homeostasis in the host. The release of endogenous mediators into the extracellular milieu by macrophages represents a practical way for macrophages to control their own activation state. These signals act in an autocrine fashion by binding to receptors on the macrophage cell surface to terminate the inflammatory process. Some of the more well-studied endogenous inhibitors include purinergic molecules, such as adenosine, and lipid mediators, including prostaglandins, resolvins, and lipoxins, among others. In this section, we review the involvement of these endogenous mediators in macrophage biology and their role in the systemic resolution of inflammation.
Purinergic Molecules
Purinergic signaling molecules, such as adenosine, are released following metabolic alterations, cell death, or tissue damage, but can also be generated in close proximity to macrophages themselves in response to infection. Adenosine is derived from adenosine triphosphate (ATP), which is generated and released by macrophages following activation. A range of stimuli, including TLRs and type I interferons, induce a metabolic switch in macrophages from oxidative phosphorylation to glycolysis, which results in the generation of intracellular ATP (53). The product of ATP catabolism, adenosine, also enhances glycolysis in TLR-activated macrophages, which suggests a positive feedback loop in the production of ATP that helps maintain the restorative functions of macrophages (54). Upon stimulation, ATP-enriched vesicles in the cytoplasm of macrophages are released by exocytosis (55). Other mechanisms of ATP release involve its conduction through pannexin-1 channels on apoptotic cells or stimulated macrophages (56).
The ATP released from macrophages is catabolized on the plasma membrane in a two-step process (57). The macrophage surface ectoenzyme CD39 (E-NTPDase1) is able to hydrolyze ATP to adenosine monophosphate (AMP) in a Ca2+- and Mg2+-dependent manner (58). Next, the surface enzyme CD73 (Ecto5 NTase) quickly converts AMP to adenosine (59). The coordinated action of these two surface enzymes leads to a rise in the concentration of adenosine directly surrounding the macrophage. Our lab recently demonstrated that this process takes place extremely rapidly, as extracellular ATP is virtually undetectable in wild-type macrophages, whereas ATP remains at relatively high levels for up to 24 h in supernatants of macrophages deficient in CD39 (57). To further highlight the importance of this catabolic process, we also demonstrated that macrophage hydrolysis of endogenous ATP via CD39 allows the cell to terminate the synthesis of inflammatory cytokines (57).
Coupled to TLR stimulation, adenosine changes the phenotype of macrophages and promotes an immunoregulatory rather than an inflammatory response (60). Macrophages respond to adenosine via four transmembrane G-protein coupled receptors, all expressed on the cell surface at different levels: A1R, A2aR, A2bR, and A3R (61). The A2a and A2b receptors are involved in promoting anti-inflammatory effects due in part to their coupling to Gαs, which leads to increases in intracellular cyclic AMP (cAMP). Macrophages stimulated with LPS selectively upregulate A2aR and A2bR (62). This upregulation enhances macrophage sensitivity to adenosine, allowing the macrophage to transition toward an immunoregulatory phenotype. The addition of adenosine to macrophages leads to the increased production of IL-10 and decreased production of TNF-α and IL-12 (57, 63). Studies using cells from knockout mice demonstrate that this suppression of TNF-α and IL-12 is enacted through both A2aR and A2bR (64, 65). The anti-inflammatory effects of adenosine are not limited to cytokine production, as studies have elucidated its role in suppressing superoxide and nitric oxide production in macrophages (66). Additionally, high-throughput RNA sequencing from our lab demonstrated that macrophages stimulated with LPS and adenosine potently downregulated many genes involved in inflammation relative to LPS alone (39).
Adenosine receptors are known to modulate a number of diseases containing inflammatory components. In vivo and in vitro studies demonstrate that adenosine promotes the resolution of tissue injury through the A2aR-specific induction of vascular endothelial growth factor production by macrophages (67). A role for adenosine was also shown in atherosclerosis, in which adenosine has widespread protective effects in the vasculature, including the inhibition of foam cell formation and reduced systemic inflammation (68). The transition of macrophages from an inflammatory to an immunoregulatory phenotype is crucial in controlling the harmful effects of endotoxemia (57).
Prostaglandin E2
Found in a wide range of tissues, prostaglandins are bioactive lipids that are implicated in many processes, including proliferation, angiogenesis, and inflammation. They are part of the prostanoid family of lipids, which are synthesized from essential fatty acids in a multistep process. First, phospholipases hydrolyze membrane phospholipids, liberating arachidonic acid from membrane lipids. Arachidonic acid is oxidized into PGG2 and reduced to PGH2 by the cyclooxygenase (COX) enzymes COX-1 and COX-2, the latter of which is highly upregulated throughout the immune system in response to proinflammatory signals, including LPS, IL-1β, and TNF-α (69). Lastly, PGH2 is converted into PGE2 via three distinct synthases: mPGES-1, mPGES-2, and cPGES. PGE2 levels are regulated by both its synthesis and degradation. 15-PGDH and 13-PGR are catabolic enzymes that can rapidly remove PGE2 from the cellular environment.
The involvement of PGE2 in acute inflammation is well documented, but paradoxically, PGE2 along with the other prostanoids PGD2 and PGJ2 also play an important role in immune suppression (70). Cells, including macrophages, sense PGE2 via four transmembrane G-protein-coupled receptors (GPCRs): EP1–4. EP2 and EP4 are coupled to Gαs proteins, which stimulate the release of intracellular cAMP. In conjunction with TLR stimulation, PGE2 suppresses IL-12 and TNF-α and partially suppresses IL-6 production in macrophages (71). At the same time, PGE2 increases IL-10 release from macrophages in a PKA-dependent manner (72). PGE2 suppresses inflammasome activation in macrophages via the EP4 receptor, attenuating the release of IL-1β (73). Additionally, IL-17 increases in the presence of PGE2, leading to the formation of M2 macrophage microenvironments (74). Our lab previously demonstrated through functional studies and high-throughput RNA sequencing that murine macrophages stimulated with LPS and PGE2 adopt an immunoregulatory phenotype (39).
PGE2 plays a role in regulating the activation of many immune cells, especially those of the innate immune system. For example, PGE2 is known to suppress natural killer cell activity by dramatically increasing intracellular cAMP (75). Lung inflammation in response to allergens and pollutants, as well as colonic inflammation, is also attenuated by PGE2 through the EP4 receptor on macrophages (76, 77). Additionally, PGE2 downregulates MHC class II expression on dendritic cells once they have migrated to lymphoid organs to suppress antigen presentation (78). A PGE2 dose dependently inhibits bacterial killing by alveolar macrophages (79). Moreover, many bacteria and intracellular parasites, such as Leishmania donovani, have developed mechanisms to induce PGE2 production by macrophages to suppress inflammation and better survive within the host (80). Mycobacterium tuberculosis (Mtb) can stimulate the production of PGE2 by macrophages through the TLR2 MAPK pathway to prevent cell necrosis (81). They also showed that EP4 antagonism in Mtb-infected macrophages inhibited the expression of COX-2 and mPGES-1, demonstrating a positive feedback loop for endogenous PGE2 production (81).
COX inhibitors are common nonsteroidal anti-inflammatory drugs (NSAIDs) used to inhibit inflammation in the host. However, chronic inhibition of COX enzymes in macrophages drives them to become more inflammatory, in part due to decreased production of PGE2 (82). Additionally, it is thought that these classic inhibitors of prostaglandin synthesis may prolong chronic inflammation when taken during the resolution stage (83).
Resolvins and Lipoxins
Similar to prostaglandins, resolvins and lipoxins are lipid mediators that prevent uncontrolled inflammation and promote the resolution of inflammation. Resolvins are a class of molecules biosynthesized from two omega-3 fatty acids: eicosapentaenoic acid and docosahexaenoic acid, leading to E resolvins (RvEs) and D resolvins (RvDs), respectively (84). RvE1 signals through chemokine receptor-like 1 and leukotriene B4 receptor, whereas RvD1 signals through GPR32 and formyl-peptide receptor (FPR2) (85). These receptors are expressed on a variety of cells, including macrophages. One function of resolvins appears to be halting the infiltration of poly-morphonuclear leukocytes to sites of infections once they are no longer needed (86). A recent study demonstrated that macrophages stimulated in the presence of resolvins, specifically RvD1, had decreased production of IL-1β, IL-6, and IL-8 (87). This was due to lower caspase-1 activation and signaling through GPR32 on the macrophage surface (87, 88). Additionally, RvD1 attenuates Th1 cytokine production by classically activated macrophages and upregulates arginase I expression (89). Both resolvins and lipoxins potently inhibit TNF production by primary human macrophages in response to purified LPS but not to live Escherichia coli (90).
Lipoxins are lipoxygenase-derived eicosanoids, and their synthesis is aspirin triggered. Both resolvins and lipoxins depend on 5-lipoxygenase-activating protein for synthesis. Two lipoxins, LXA4 and LXB4, are derived from arachidonic acid following an inflammatory signal. Lipoxins are able to interact with a number of receptors on the cell surface, including leukotriene receptor CysLT1 and the N-formyl peptide receptor FPR2. Lipoxins enhance the phagocytosis of apoptotic and necrotic cells and are important components of the resolution process.
The production of endogenous molecules that contain resolving properties is critical for the macrophage to control its activation. Adenosine, prostaglandins, and the group of lipid mediators discussed above all have one commonality: They are sensed by GPCRs on the cell surface. GPCRs have long been of interest in drug development due to their widespread expression and therapeutic potential. GPCRs have profound effects on regulating macrophage function through the actions of cAMP and Ca2+ (91). Luckily, many tools are available for the manipulation of these receptors to study their roles in inflammation and its resolution. Scholars have long thought that the return to homeostasis by macrophages is a passive process, but in the following section we try to illuminate the active and programmed mechanisms that macrophages initiate to control their activation state.
THE REGULATION OF MACROPHAGES BY EXOGENOUS INHIBITORS
The regulation of macrophage inflammatory responses can occur through the autocrine mechanisms described above, but it can also occur via the addition of numerous exogenous signals to the macrophage microenvironment. Some of these signals turn off inflammatory responses, some turn on anti-inflammatory responses, and some of these signals result in cell death to terminate inflammatory responses.
Turning Off Inflammation
The growing base of knowledge regarding the synthesis and control of inflammatory mediators is leading to the development of inhibitors of these molecular responses. For example, the central role that TNF-α plays in initiating the inflammatory cascade has led to the remarkably successful development of humanized monoclonal antibodies to TNF-α to treat inflammatory diseases (92). Similarly, our improved understanding of the role of inflammasomes in cytosolic host defense has led to the development of drugs to target IL-1β, its receptors, and the host molecules responsible for the secretion of IL-1β and IL-18 (93). Other approaches to drug development are less specific and generally try to inhibit inflammation by targeting the production of inflammatory cytokines or lipid mediators in general. As described above, during inflammation, phospholipase A2 becomes activated and membrane phospholipids release arachidonic acid to promote eicosanoid production (primarily prostaglandins, leukotrienes, and thromboxane). NSAIDs were among the earliest synthetic inhibitors of inflammation, which generally work by inhibiting COX enzymes and reducing eicosanoid production to counteract situations in which prostaglandins might be inflammatory (82).
Glucocorticoids are also a general class of macrophage modulators that can reduce the production of a number of inflammatory cytokines, including TNF-α, IL-1β, IL-6, and IL-12, by activated macrophages (94). Glucocorticoid receptors (GRs) have multiple modes of action that help reduce inflammation. First, the complex of ligand and receptor can migrate into the nucleus where it binds to specific response elements. This leads to chromatin remodeling that can initiate or inhibit transcriptional machinery (94). Interference of NF-κB and AP-1 activity, among other inflammatory mediators by GRs, can also occur through direct physical interaction (95). More rapid effects of glucocorticoids include activation of or interactions with MAPKs, adenylyl cyclase, protein kinase C, and heterotrimeric guanosine triphosphate-binding proteins (96). Glucocorticoids also induce synthesis of IκB, which compensates for its degradation during inflammation, decreasing the amount of NF-κB that translocates into the nucleus (97). Through these mechanisms, glucocorticoids inhibit the transcription of proinflammatory cytokines, reduce antigen presentation, and decrease mRNA stability, while increasing the phagocytosis of apoptotic cells leading to anti-inflammatory transforming growth factor-beta (TGF-β) production by macrophages (60, 98). Exogenous addition of naturally made steroid hormones such as glucocorticoids has proven to be effective in the treatment of autoimmune diseases such as MS, rheumatoid arthritis, psoriasis, and many more (94).
Kinase Inhibitors
The initiation of inflammatory pathways following receptor ligation relies heavily on the activation of kinase cascades. General and specific inhibition of such kinases is a widely applied field, focused mainly on the Janus activated kinases (JAKs), spleen tyrosine kinase (SYK), and the MAPKs. All are involved in signaling via TLRs and many cytokine receptors. The JAK/STAT pathway initiates cytokine receptor signaling by phosphorylating tyrosine residues on the receptor and on STATs, activating downstream immune response genes. Interruption of JAK activity prevents the production of IFN-γ and IL-6 and has proven to be effective in treating arthritis, ulcerative colitis, and psoriasis (99). By associating with receptors containing immunoreceptor tyrosine-based activation motifs, SYK plays an important role in a growing number of immune pathways, including cellular adhesion, pathogen recognition, and inflammasome activation (100). Inhibition of this kinase reduces inflammatory degradation of bone in rheumatoid arthritis (101). Research in various disease models shows reduced inflammatory pathology due to SYK inhibition. For example, in rheumatoid arthritis patients, SYK inhibitors demonstrated a reduction in inflammatory IL-6 and MMP-3, whereas SYK inhibition in mast cells prevented degranulation via IgE receptor signaling (100). The well-conserved MAPKs play an important role in proinflammatory signaling cascades, especially p38 kinases and Jun amino-terminal kinases (JNKs) (102). Phosphorylation of JNKs leads to increased c-Jun activation, an important component of the AP-1 complex involved in cytokine gene expression (103). The p38 MAPKs regulate expression of IL-1β, TNF-α, IL-6, IL-8, and even COX-2 (104). In the Crohn’s disease model, inhibition of both MAPKs resulted in the improved clinical status of patients (105). The development of specific inhibitors of these kinases continues to show great promise in the regulation of inflammatory responses.
Receptor-Mediated Inhibition
Just as many pattern recognition receptors on macrophages can induce inflammatory responses, many macrophages can initiate inhibitory signals to counter macrophage activation and reestablish homeostasis. Signaling through these receptors can reduce inflammatory cytokine production and inhibit free radical production. The signal regulatory protein α on macrophages, acting through tyrosine-based inhibitory motifs, binds CD47 and produces negative signals to prevent phagocytic activity, the production of superoxide, and inflammatory cytokines. This process is mediated through the activation of SHP-1 and SHP-2, phosphatases that counteract kinase activity (106). Similarly, a variety of cell types express CD200, and its interaction with CD200R on myeloid cells produces inhibitory signals. This interaction serves as a constitutive restraint on inflammatory stimulation of macrophages, limiting autoimmune pathology (107). Acting through the downstream kinase, CD200R signaling inhibits Ras, subsequently reducing PI3K signals and inhibiting cytokine production (108). The engagement of CD200 to its receptor leads to reduced responses to IFN-γ and IL-17 stimulation in mouse peritoneal macrophages and decreased IL-5 and IL-13 production following tetanus toxoid stimulation in human peripheral blood mononuclear cells (109). Receptors such as TREM2 can also regulate inflammation. The expression of TREM2 on recently differentiated macrophages entering into sites of inflammation acts in cooperation with DAP-12 to reduce macrophage production of TNF-α and IL-6 (110). The ligands for TREM2 are still under investigation but include endogenously produced molecules and also exogenous compounds such as LPS (111). These and other receptors share the common goal of preventing the overproduction of inflammatory signals by macrophages.
Finally, naturally produced molecules at a given anatomical site can locally dampen macrophage inflammatory responses. For example, intestinal mucosal surfaces help to protect epithelial cells against potentially harmful contents within the intestinal lumen, using phospholipids to establish a hydrophobic surface that can inhibit excess inflammation (112). Phosphatidylcholine, one of the most prevalent phospholipids in the mucosa, can inhibit kinase activation, NF-κB transport, and proinflammatory gene production. Similarly, pulmonary surfactants play an important role in reducing inflammatory responses in the lungs (113). Phosphatidylcholine also decreases phagosome assembly and the killing of Mtb, an infection requiring Th1 inflammatory responses for clearance (112).
Turning On Anti-Inflammation
The resolution of inflammatory responses by macrophages is not simply dependent on turning off inflammatory signal production but also by increasing the production of anti-inflammatory mediators. The most common of these mediators are IL-10 and TGF-β. IL-10 signals through the IL-10R on macrophages, inhibiting inflammatory cytokine production and frequently decreasing the killing of intracellular pathogens (114). Many cells produce IL-10, and genetic alterations that result in a failure to produce this cytokine are invariably associated with inflammatory im-munopathology. However, the potent immunoregulatory activity of IL-10, which was repeatedly observed by many investigators using a number of inflammatory model systems, has generally not led to the utilization of recombinant IL-10 to treat autoimmunity. Unfortunately, the administration of recombinant IL-10 is typically not able to substantially reverse inflammatory pathology in patients suffering with autoimmunity, including psoriasis and Crohn’s disease (115). One reason may be that IL-10 is frequently diverted to the many different cells that have receptors for IL-10, preventing its delivery to inflamed areas. The local induction of IL-10 may be more effective in regulating inflammation. Whether produced by macrophages or other cell types, IL-10 release depends on the strength of stimulation (115). In macrophages, increased signaling that occurs through a combination of stimuli results in high levels of IL-10 production. For example, the addition of LPS in combination with the ligation of macrophage Fcγ receptors (FcγRs) by high-density immune complexes can potently downregulate IL-12 production and induce high levels of IL-10 production (116). This reciprocal alteration in these two key cytokines results in a macrophage population that can mitigate inflammation and provide protection against acute endo-toxicity. These macrophages also exhibit increased susceptibility to intracellular pathogens (117). Inflammation induced by a variety of stimuli, including virtually all TLR ligands, CD40:CD40L, and low molecular weight hyaluronic acid, was modulated when coupled with macrophage FcγR ligation. The physiological relevance of this phenotypic alteration was demonstrated in an infectious disease model in which the intracellular parasite Leishmania sp. uses this pathway to induce macrophage IL-10 to promote its survival within macrophages (117).
Although TGF-β exhibits a wider range of roles in cellular processes when compared to IL-10, its ability to modulate inflammation and influence disease progression renders it an extremely important immune modulator. The abundance and activity of TGF-β ligands, the presence of SMAD cofactors, and epigenetic modifications all contribute to the intensity and type of signal created from TGF-β signaling (118). SMAD proteins, activated via the TGF-β receptor, form complexes with each other and with other cofactors to activate or inhibit gene transcription. The addition of exogenous TGF-β to LPS-stimulated macrophages decreased the release of proinflammatory cytokines, suggesting paracrine functions of the signaling (119). In mouse models, TGF-β treatment prevented disease in collagen-induced arthritis and prevented relapse in rheumatoid arthritis, consistent with its anti-inflammatory activity (120). Use of TGF-β as a therapeutic in mouse models for multiple sclerosis (i.e., EAE) showed improved clinical status and decreased nervous system damage, indicating its promise (121). However, the multifaceted effect of TGF-β production is illustrated by its contribution to fibrotic diseases, showing the need to continue studying this complex pathway (122).
Although the functional regulation of the inflammatory response is important in homeostatic maintenance, numerous organisms exploit these pathways to establish a successful infection and to survive in macrophages. Leishmania spp. can interfere with MAPK signaling to decrease IL-12 production and promote IL-10 production (123). M. tuberculosis relies on an inhibition of IFN-γ and promotion of IL-10 production to survive and replicate in macrophages (124). Intestinal and urogenital bacteria such as Lactobacillus rhamnosus can promote anti-inflammatory responses from macrophages. The commensal bacteria’s secretome was able to induce granulocyte colony-stimulating factor production by macrophages, leading to STAT3 activation, JNK inactivation, and TNF-α suppression in LPS- or E. coli-activated macrophages (125). Various nematodes and helminths can powerfully regulate the immune response, decreasing inflammatory Th1/Th17 responses and promoting Th2 response environments (126).
Cell Death and Clearance
Cell death can contribute to a decrease in inflammation. The induction of apoptosis is an important process used to remove inflammatory macrophages (127). The rapid removal of macrophages helps to prevent chronic inflammation and decrease fibrosis. In the lungs, the macrophages recruited to the site of inflammation exhibit high levels of Fas, and the induction of apoptosis via Fas-activating antibodies leads to clearance (128). Endoplasmic reticulum stress in combination with pattern recognition receptor activation may also contribute to macrophage apoptosis (129). Additional mechanisms of macrophage cell death and the importance of these processes in the regulation of inflammation are important research areas that remain under investigation.
The clearance of dead or dying cells by macrophages is a central role of these cells. The uptake of apoptotic cells by macrophage is characterized, and the link to anti-inflammatory responses is well described. Generally, macrophage-mediated clearance of apoptotic cells induces the production of lipoxins, resolvins, and protectins that work in autocrine and paracrine fashion to promote the resolution of inflammation (98). During the immune response, one of the most commonly phagocytized apoptotic cells is the neutrophil. Neutrophils are among the first responders to pathogens, and the potentially toxic molecules released by neutrophils must be tightly regulated. Their clearance subsequent to apoptosis is mediated primarily by macrophages that recognize specific changes on the cell surface (130). Recognition of the common apoptotic marker phosphatidylserine promotes TGF-β production by macrophages to limit inflammation (131). Annexin A1 is exported by apoptotic and necrotic cells. It can bind to phosphatidylserine on the outer membrane, helping to enhance phagocytic uptake (132). Apoptotic cell clearance by macrophages is especially important in the continuous process of cellular turnover as organisms develop and grow (98).
MACROPHAGE ACTIVATION SYNDROME: A FAILURE TO REGULATE
Macrophage activation syndrome (MAS) is a relatively rare autoimmune complication that can follow chronic infectious diseases, T cell immunotherapy, or rheumatologic diseases, especially in children. Persistent fever, coagulopathies, lymphadenopathy, and hepatosplenomegaly are frequently associated with this disease, and hemophagocytosis and hyperferritinemia are striking features of MAS (133). The secretion of IFN-γ seems to be an early event in this disease that leads to a cytokine storm characterized by myriad proinflammatory cytokines and chemokines from macrophages, including IL-1β, IL-6, and TNF-α (134). A central role for activated T cells, especially CD8 T cells, is generally accepted, and in humans there is a strong correlation between IFN-γ production from these cells and clinical pathology. In experimental animal models, recurrent TLR activation has been used to induce disease (135), but in humans there is also strong evidence for the activation of inflammasomes, including NLRC4, in this disease (136). The use of anakinra (an IL-1 receptor antagonist) to treat a subset of patients with MAS supports a role for macrophage-derived IL-1 in disease pathology (137). IL-18 is another cytokine that depends on caspase 1 cleavage for secretion, and elevated IL-18 may contribute to MAS pathogenesis, perhaps by inducing additional IFN-γ production (138). One interesting hypothesis is that the failure of activated macrophages to die in a timely manner is in part responsible for this autoinflammatory syndrome. Indeed, this disease and the closely related hemophagic lymphohistiocytosis are associated with mutations that interfere with efficient cytotoxicity (133). The inability of cytotoxic cells to induce apoptosis of activated macrophages may allow these cells to persist and produce inflammatory mediators for prolonged periods of time.
The first line treatment for MAS is corticosteroids, with cyclosporine added if necessary. High-dose intravenous immunoglobulin in combination with corticosteroids is also used as a treatment option when corticosteroids alone prove to be insufficient. Humanized monoclonal antibodies to IL-1 or IL-6 are now frequently added. Because of the accepted practice of treating this disease with drugs that target macrophage cytokine production, one can consider MAS in the simplest terms to be a failure to efficiently regulate macrophage activation in response to inflammatory stimulation. MAS illustrates the enormous pathological potential of activated macrophages and the pressing need to develop new ways of regulating macrophage activation. Current therapies are largely directed at blocking inflammatory cytokine production or preventing the binding of cytokines to their cognate receptors. This therapeutic approach transiently diminishes clinical symptomatology but does not reverse disease or provide long-term protection from recurrences. New ways are needed to reprogram macrophages to change their activation state or to educate them to produce immunoregulatory molecules rather than immunostimulatory molecules.
SUMMARY
Although macrophages represent a potentially lethal source of inflammatory cytokines, they are floating in a sea of regulators that can dampen inflammation and prevent autoimmune sequelae. These regulators prevent cytokine overproduction by a variety of different mechanisms. They include transcriptional repressors, epigenetic silencers, inhibitors of signaling kinases, mechanisms to degrade signaling molecules, and small regulatory RNAs. All of these regulators work to maintain the delicate balance of a healthy homeostatic system of host defense.
Acknowledgments
This study was funded by a grant from the National Institutes of Health (R01 GM102589-01).
Footnotes
DISCLOSURE STATEMENT
The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.
LITERATURE CITED
- 1.Medzhitov R, Horng T. Transcriptional control of the inflammatory response. Nat Rev Immunol. 2009;9:692–703. doi: 10.1038/nri2634. [DOI] [PubMed] [Google Scholar]
- 2.Hargreaves DC, Horng T, Medzhitov R. Control of inducible gene expression by signal-dependent transcriptional elongation. Cell. 2009;138:129–45. doi: 10.1016/j.cell.2009.05.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Escoubet-Lozach L, Benner C, Kaikkonen MU, Lozach J, Heinz S, et al. Mechanisms establishing TLR4-responsive activation states of inflammatory response genes. PLOS Genet. 2011;7:e1002401. doi: 10.1371/journal.pgen.1002401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Barozzi I, Simonatto M, Bonifacio S, Yang L, Rohs R, et al. Co-regulation of transcription factor binding and nucleosome occupancy through DNA features of mammalian enhancers. Mol Cell. 2014;54:844–57. doi: 10.1016/j.molcel.2014.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ghisletti S, Barozzi I, Mietton F, Polletti S, De Santa F, et al. Identification and characterization of enhancers controlling the inflammatory gene expression program in macrophages. Immunity. 2010;32:317–28. doi: 10.1016/j.immuni.2010.02.008. [DOI] [PubMed] [Google Scholar]
- 6.Boehm U, Klamp T, Groot M, Howard JC. Cellular responses to interferon-γ. Annu Rev Immunol. 1997;15:749–95. doi: 10.1146/annurev.immunol.15.1.749. [DOI] [PubMed] [Google Scholar]
- 7.Qiao Y, Giannopoulou EG, Chan CH, Park S, Gong S, et al. Synergistic activation of inflammatory cytokine genes by interferon-γ-induced chromatin remodeling and Toll-like receptor signaling. Immunity. 2013;39:454–69. doi: 10.1016/j.immuni.2013.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bergeron M, Olivier M. Trypanosoma cruzi-mediated IFN-γ-inducible nitric oxide output in macrophages is regulated by iNOS mRNA stability. J Immunol. 2006;177:6271–80. doi: 10.4049/jimmunol.177.9.6271. [DOI] [PubMed] [Google Scholar]
- 9.Su X, Yu Y, Zhong Y, Giannopoulou EG, Hu X, et al. Interferon-γ regulates cellular metabolism and mRNA translation to potentiate macrophage activation. Nat Immunol. 2015;16:838–49. doi: 10.1038/ni.3205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Fukao T, Tanabe M, Terauchi Y, Ota T, Matsuda S, et al. PI3K-mediated negative feedback regulation of IL-12 production in DCs. Nat Immunol. 2002;3:875–81. doi: 10.1038/ni825. [DOI] [PubMed] [Google Scholar]
- 11.Martin M, Rehani K, Jope RS, Michalek SM. Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol. 2005;6:777–84. doi: 10.1038/ni1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Heremans H, Van Damme J, Dillen C, Dijkmans R, Billiau A. Interferon gamma, a mediator of lethal lipopolysaccharide-induced Shwartzman-like shock reactions in mice. J Exp Med. 1990;171:1853–69. doi: 10.1084/jem.171.6.1853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kühbacher T, Gionchetti P, Hampe J, Helwig U, Rosenstiel P, et al. Activation of signal-transducer and activator of transcription 1 (STAT1) in pouchitis. Clin Exp Immunol. 2001;123:395–401. doi: 10.1046/j.1365-2249.2001.01455.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, et al. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. PNAS. 2003;100:2610–15. doi: 10.1073/pnas.0337679100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Uzel G, Sampaio EP, Lawrence MG, Hsu AP, Hackett M, et al. Dominant gain-of-function STAT1 mutations in FOXP3 wild-type immune dysregulation–polyendochrinopathy–enteropathy–X-linked-like syndrome. J Allergy Clin Immunol. 2013;131:1611–23.e3. doi: 10.1016/j.jaci.2012.11.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Rosenzweig SD, Holland SM. Defects in the interferon-γ and interleukin-12 pathways. Immunol Rev. 2005;203:38–47. doi: 10.1111/j.0105-2896.2005.00227.x. [DOI] [PubMed] [Google Scholar]
- 17.van der Pouw Kraan TCTM, van Gaalen FA, Kasperkovitz PV, Verbeet NL, Smeets TJM, et al. Rheumatoid arthritis is a heterogeneous disease: evidence for differences in the activation of the STAT-1 pathway between rheumatoid tissues. Arthritis Rheum. 2003;48:2132–45. doi: 10.1002/art.11096. [DOI] [PubMed] [Google Scholar]
- 18.Toro JR, Finlay D, Dou X, Zheng SC, Le Boit PE, Connolly MK. Detection of type 1 cytokines in discoid lupus erythematosus. Arch Dermatol. 2000;136:1497–501. doi: 10.1001/archderm.136.12.1497. [DOI] [PubMed] [Google Scholar]
- 19.Browne TC, McQuillan K, McManus RM, O’Reilly J-A, Mills KHG, Lynch MA. IFN-γ production by amyloid β-specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer’s disease. J Immunol. 2013;190:2241–51. doi: 10.4049/jimmunol.1200947. [DOI] [PubMed] [Google Scholar]
- 20.Kryczek I, Bruce AT, Gudjonsson JE, Johnston A, Aphale A, et al. Induction of IL-17+ T cell trafficking and development by IFN-γ: mechanism and pathological relevance in psoriasis. J Immunol. 2008;181:4733–41. doi: 10.4049/jimmunol.181.7.4733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Arellano G, Ottum PA, Reyes LI, Burgos PI, Naves R. Stage-specific role of interferon-gamma in experimental autoimmune encephalomyelitis and multiple sclerosis. Front Immunol. 2015;6:492. doi: 10.3389/fimmu.2015.00492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kebir H, Ifergan I, Alvarez JI, Bernard M, Poirier J, et al. Preferential recruitment of interferon-γ-expressing Th17 cells in multiple sclerosis. Ann Neurol. 2009;66:390–402. doi: 10.1002/ana.21748. [DOI] [PubMed] [Google Scholar]
- 23.Kallaur AP, Oliveira SR, Simão ANC, Alfieri DF, Flauzino T, et al. Cytokine profile in patients with progressive multiple sclerosis and its association with disease progression and disability. Mol Neurobiol. 2016 doi: 10.1007/s12035-016-9846-x. In press. [DOI] [PubMed] [Google Scholar]
- 24.Hindinger C, Bergmann CC, Hinton DR, Phares TW, Parra GI, et al. IFN-γ signaling to astrocytes protects from autoimmune mediated neurological disability. PLOS ONE. 2012;7:e42088. doi: 10.1371/journal.pone.0042088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Divanovic S, Trompette A, Atabani SF, Madan R, Golenbock DT, et al. Negative regulation of Toll-like receptor 4 signaling by the Toll-like receptor homolog RP105. Nat Immunol. 2005;6:571–78. doi: 10.1038/ni1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.O’Neill LAJ, Bowie AG. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol. 2007;7:353–64. doi: 10.1038/nri2079. [DOI] [PubMed] [Google Scholar]
- 27.Janssens S, Burns K, Vercammen E, Tschopp J, Beyaert R. Myd88S, a splice variant of Myd88, differentially modulates NF-κB- and AP-1-dependent gene expression. FEBS Lett. 2003;548:103–7. doi: 10.1016/s0014-5793(03)00747-6. [DOI] [PubMed] [Google Scholar]
- 28.Palsson-McDermott EM, Doyle SL, McGettrick AF, Hardy M, Husebye H, et al. TAG, a splice variant of the adaptor TRAM, negatively regulates the adaptor Myd88-independent TLR4 pathway. Nat Immunol. 2009;10:579–86. doi: 10.1038/ni.1727. [DOI] [PubMed] [Google Scholar]
- 29.Wang Y, Tang Y, Teng L, Wu Y, Zhao X, Pei G. Association of β-arrestin and TRAF6 negatively regulates Toll-like receptor-interleukin 1 receptor signaling. Nat Immunol. 2006;7:139–47. doi: 10.1038/ni1294. [DOI] [PubMed] [Google Scholar]
- 30.Boone DL, Turer EE, Lee EG, Ahmad R-C, Wheeler MT, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol. 2004;5:1052–60. doi: 10.1038/ni1110. [DOI] [PubMed] [Google Scholar]
- 31.Hayden MS, Ghosh S. Shared principles in NF-κB signaling. Cell. 2008;132:344–62. doi: 10.1016/j.cell.2008.01.020. [DOI] [PubMed] [Google Scholar]
- 32.Barish GD, Yu RT, Karunasiri M, Ocampo CB, Dixon J, et al. Bcl-6 and NF-κB cistromes mediate opposing regulation of the innate immune response. Genes Dev. 2010;24:2760–65. doi: 10.1101/gad.1998010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Toney LM, Cattoretti G, Graf JA, Merghoub T, Pandolfi P-P, et al. BCL-6 regulates chemokine gene transcription in macrophages. Nat Immunol. 2000;1:214–20. doi: 10.1038/79749. [DOI] [PubMed] [Google Scholar]
- 34.Maine GN, Mao X, Komarck CM, Burstein E. COMMD1 promotes the ubiquitination of NF-κB subunits through a cullin-containing ubiquitin ligase. EMBO J. 2007;26:436–47. doi: 10.1038/sj.emboj.7601489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Labzin LI, Schmidt SV, Masters SL, Beyer M, Krebs W, et al. ATF3 is a key regulator of macrophage IFN responses. J Immunol. 2015;195:4446–55. doi: 10.4049/jimmunol.1500204. [DOI] [PubMed] [Google Scholar]
- 36.Hutchins AP, Diez D, Miranda-Saavedra D. The IL-10/STAT3-mediated anti-inflammatory response: recent developments and future challenges. Brief Funct Genom. 2013;12:489–98. doi: 10.1093/bfgp/elt028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bode JG, Ehlting C, Haussinger D. The macrophage response towards LPS and its control through the P38MAPK-STAT3 axis. Cell Signal. 2012;24:1185–94. doi: 10.1016/j.cellsig.2012.01.018. [DOI] [PubMed] [Google Scholar]
- 38.Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, et al. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med. 2007;357:1608–19. doi: 10.1056/NEJMoa073687. [DOI] [PubMed] [Google Scholar]
- 39.Fleming BD, Chandrasekaran P, Dillon LAL, Dalby E, Suresh R, et al. The generation of macrophages with anti-inflammatory activity in the absence of STAT6 signaling. J Leukoc Biol. 2015;98:395–407. doi: 10.1189/jlb.2A1114-560R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Investig. 2012;122:787–95. doi: 10.1172/JCI59643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Huber S, Hoffmann R, Muskens F, Voehringer D. Alternatively activated macrophages inhibit T-cell proliferation by STAT6-dependent expression of PD-L2. Blood. 2010;116:3311–20. doi: 10.1182/blood-2010-02-271981. [DOI] [PubMed] [Google Scholar]
- 42.Hesse M, Modolell M, La Flamme AC, Schito M, Fuentes JM, et al. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism. J Immunol. 2001;167:6533–44. doi: 10.4049/jimmunol.167.11.6533. [DOI] [PubMed] [Google Scholar]
- 43.Zhang Y, Zhang M, Zhong M, Suo Q, Lv K. Expression profiles of miRNAs in polarized macrophages. Int J Mol Med. 2013;31:797–802. doi: 10.3892/ijmm.2013.1260. [DOI] [PubMed] [Google Scholar]
- 44.Graff JW, Dickson AM, Clay G, McCaffrey AP, Wilson ME. Identifying functional microRNAs in macrophages with polarized phenotypes. J Biol Chem. 2012;287:21816–25. doi: 10.1074/jbc.M111.327031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ponomarev ED, Veremeyko T, Barteneva N, Krichevsky AM, Weiner HL. MicroRNA-124 promotes microglia quiescence and suppresses EAE by deactivating macrophages via the C/EBP-α-PU.1 pathway. Nat Med. 2011;17:64–70. doi: 10.1038/nm.2266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Veremeyko T, Siddiqui S, Sotnikov I, Yung A, Ponomarev ED. IL-4/IL-13-dependent and independent expression of miR-124 and its contribution to M2 phenotype of monocytic cells in normal conditions and during allergic inflammation. PLOS ONE. 2013;8:e81774. doi: 10.1371/journal.pone.0081774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wu X, Dai Y, Yang Y, Huang C, Meng X, et al. Emerging role of microRNAs in regulating macrophage activation and polarization in immune response and inflammation. Immunology. 2016;148:237–48. doi: 10.1111/imm.12608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Curtale G, Mirolo M, Renzi TA, Rossato M, Bazzoni F, Locati M. Negative regulation of Toll-like receptor 4 signaling by IL-10-dependent microRNA-146b. PNAS. 2013;110:11499–504. doi: 10.1073/pnas.1219852110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhang Y, Zhang H, Liu Z. MicroRNA-147 suppresses proliferation, invasion and migration through the AKT/MTOR signaling pathway in breast cancer. Oncol Lett. 2016;11:405–10. doi: 10.3892/ol.2015.3842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Rossato M, Curtale G, Tamassia N, Castellucci M, Mori L, et al. IL-10-induced microRNA-187 negatively regulates TNF-α, IL-6, and IL-12p40 production in TLR4-stimulated monocytes. PNAS. 2012;109:E3101–10. doi: 10.1073/pnas.1209100109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Suojalehto H, Lindström I, Majuri M-L, Mitts C, Karjalainen J, et al. Altered microRNA expression of nasal mucosa in long-term asthma and allergic rhinitis. Int Arch Allergy Immunol. 2014;163:168–78. doi: 10.1159/000358486. [DOI] [PubMed] [Google Scholar]
- 52.Rückerl D, Jenkins SJ, Laqtom NN, Gallagher IJ, Sutherland TE, et al. Induction of IL-4rα-dependent microRNAs identifies PI3K/AKT signaling as essential for IL-4-driven murine macrophage proliferation in vivo. Blood. 2012;120:2307–16. doi: 10.1182/blood-2012-02-408252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kelly B, O’Neill LAJ. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25:771–84. doi: 10.1038/cr.2015.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ruiz-García A, Monsalve E, Novellasdemunt L, Navarro-Sabaté À, Manzano A, et al. Cooperation of adenosine with macrophage Toll-4 receptor agonists leads to increased glycolytic flux through the enhanced expression of PFKFB3 gene. J Biol Chem. 2011;286:19247–58. doi: 10.1074/jbc.M110.190298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sakaki H, Tsukimoto M, Harada H, Moriyama Y, Kojima S. Autocrine regulation of macrophage activation via exocytosis of ATP and activation of P2Y11 receptor. PLOS ONE. 2013;8:e59778. doi: 10.1371/journal.pone.0059778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chekeni FB, Elliott MR, Sandilos JK, Walk SF, Kinchen JM, et al. Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis. Nature. 2010;467:863–67. doi: 10.1038/nature09413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cohen HB, Briggs KT, Marino JP, Ravid K, Robson SC, Mosser DM. TLR stimulation initiates a CD39-based autoregulatory mechanism that limits macrophage inflammatory responses. Blood. 2013;122:1935–45. doi: 10.1182/blood-2013-04-496216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kaczmarek E, Koziak K, Sévigny J, Siegel JB, Anrather J, et al. Identification and characterization of CD39/vascular ATP diphosphohydrolase. J Biol Chem. 1996;271:33116–22. doi: 10.1074/jbc.271.51.33116. [DOI] [PubMed] [Google Scholar]
- 59.Zimmermann H, Braun N. Ecto-nucleotidases—molecular structures, catalytic properties, and functional roles in the nervous system. Prog Brain Res. 1999;120:371–85. [PubMed] [Google Scholar]
- 60.Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–69. doi: 10.1038/nri2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Haskó G, Cronstein B. Regulation of inflammation by adenosine. Front Immunol. 2013;4:85. doi: 10.3389/fimmu.2013.00085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cohen HB, Ward A, Hamidzadeh K, Ravid K, Mosser DM. IFN-γ prevents adenosine receptor (A2bR) upregulation to sustain the macrophage activation response. J Immunol. 2015;195:3828–37. doi: 10.4049/jimmunol.1501139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Németh ZH, Lutz CS, Csóka B, Deitch EA, Leibovich SJ, et al. Adenosine augments IL-10 production by macrophages through an A2B receptor-mediated posttranscriptional mechanism. J Immunol. 2005;175:8260–70. doi: 10.4049/jimmunol.175.12.8260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kreckler LM, Wan TC, Ge Z-D, Auchampach JA. Adenosine inhibits tumor necrosis factor-α release from mouse peritoneal macrophages via A2A and A2B but not the A3 adenosine receptor. J Pharmacol Exp Ther. 2006;317:172–80. doi: 10.1124/jpet.105.096016. [DOI] [PubMed] [Google Scholar]
- 65.Sun Y, Duan Y, Eisenstein AS, Hu W, Quintana A, et al. A novel mechanism of control of NFκB activation and inflammation involving A2B adenosine receptors. J Cell Sci. 2012;125:4507–17. doi: 10.1242/jcs.105023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Haskó G, Szabó C, Németh ZH, Kvetan V, Pastores SM, Vizi ES. Adenosine receptor agonists differentially regulate IL-10, TNF-α, and nitric oxide production in raw 264.7 macrophages and in endotoxemic mice. J Immunol. 1996;157:4634–40. [PubMed] [Google Scholar]
- 67.Leibovich SJ, Chen J-F, Pinhal-Enfield G, Belem PC, Elson G, et al. Synergistic up-regulation of vascular endothelial growth factor expression in murine macrophages by adenosine A2A receptor agonists and endotoxin. Am J Pathol. 2002;160:2231–44. doi: 10.1016/S0002-9440(10)61170-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Koupenova M, Johnston-Cox H, Ravid K. Regulation of atherosclerosis and associated risk factors by adenosine and adenosine receptors. Curr Atheroscler Rep. 2012;14:460–68. doi: 10.1007/s11883-012-0263-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Park JY, Pillinger MH, Abramson SB. Prostaglandin E2 synthesis and secretion: the role of PGE2 synthases. Clin Immunol. 2006;119:229–40. doi: 10.1016/j.clim.2006.01.016. [DOI] [PubMed] [Google Scholar]
- 70.Kalinski P. Regulation of immune responses by prostaglandin E2. J Immunol. 2012;188:21–28. doi: 10.4049/jimmunol.1101029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kuroda E, Yamashita U. Mechanisms of enhanced macrophage-mediated prostaglandin E2 production and its suppressive role in Th1 activation in Th2-dominant BALB/c mice. J Immunol. 2003;170:757–64. doi: 10.4049/jimmunol.170.2.757. [DOI] [PubMed] [Google Scholar]
- 72.MacKenzie KF, Clark K, Naqvi S, McGuire VA, Nöehren G, et al. PGE2 induces macrophage IL-10 production and a regulatory-like phenotype via a protein kinase A-SIK-CRTC3 pathway. J Immunol. 2013;190:565–77. doi: 10.4049/jimmunol.1202462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Sokolowska M, Chen L-Y, Liu Y, Martinez-Anton A, Qi H-Y, et al. Prostaglandin E2 inhibits NLRP3 inflammasome activation through EP4 receptor and intracellular cyclic AMP in human macrophages. J Immunol. 2015;194:5472–87. doi: 10.4049/jimmunol.1401343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Liu L, Ge D, Ma L, Mei J, Liu S, et al. Interleulin-17 and prostaglandin E2 are involved in formation of an M2 macrophage-dominant microenvironment in lung cancer. J Thorac Oncol. 2012;7:1091–100. doi: 10.1097/JTO.0b013e3182542752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Goto T, Herberman RB, Maluish A, Strong DM. Cyclic AMP as a mediator of prostaglandin E-induced suppression of human natural killer cell activity. J Immunol. 1983;130:1350–55. [PubMed] [Google Scholar]
- 76.Birrell MA, Maher SA, Dekkak B, Jones V, Wong S, et al. Anti-inflammatory effects of PGE2 in the lung: role of the EP4 receptor subtype. Thorax. 2015;70:740–47. doi: 10.1136/thoraxjnl-2014-206592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Nakatsuji M, Minami M, Seno H, Yasui M, Komekado H, et al. EP4 receptor-associated protein in macrophages ameliorates colitis and colitis-associated tumorigenesis. PLOS Genet. 2015;11:e1005542. doi: 10.1371/journal.pgen.1005542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Harizi H, Juzan M, Grosset C, Rashedi M, Gualde N. Dendritic cells issued in vitro from bone marrow produce PGE2 that contributes to the immunomodulation induced by antigen-presenting cells. Cell Immunol. 2001;209:19–28. doi: 10.1006/cimm.2001.1785. [DOI] [PubMed] [Google Scholar]
- 79.Serezani CH, Chung J, Ballinger MN, Moore BB, Aronoff DM, Peters-Golden M. Prostaglandin E2 suppresses bacterial killing in alveolar macrophages by inhibiting NADPH oxidase. Am J Respir Cell Mol Biol. 2007;37:562–70. doi: 10.1165/rcmb.2007-0153OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Saha A, Biswas A, Srivastav S, Mukherjee M, Das PK, Ukil A. Prostaglandin E2 negatively regulates the production of inflammatory cytokines/chemokines and IL-17 in visceral leishmaniasis. J Immunol. 2014;193:2330–39. doi: 10.4049/jimmunol.1400399. [DOI] [PubMed] [Google Scholar]
- 81.Nishimura T, Zhao X, Gan H, Koyasu S, Remold HG. The prostaglandin E2 receptor EP4 is integral to a positive feedback loop for prostaglandin E2 production in human macrophages infected with Mycobacterium tuberculosis. FASEB J. 2013;27:3827–36. doi: 10.1096/fj.13-228858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Na YR, Yoon YN, Son D, Jung D, Gu GJ, Seok SH. Consistent inhibition of cyclooxygenase drives macrophages towards the inflammatory phenotype. PLOS ONE. 2015;10:e0118203. doi: 10.1371/journal.pone.0118203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Scher JU, Pillinger MH. The anti-inflammatory effects of prostaglandins. J Investig Med. 2015;57:703–8. doi: 10.2310/JIM.0b013e31819aaa76. [DOI] [PubMed] [Google Scholar]
- 84.Serhan CN, Chiang N, Dalli J. The resolution code of acute inflammation: novel pro-resolving lipid mediators in resolution. Semin Immunol. 2015;27:200–15. doi: 10.1016/j.smim.2015.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Qu Q, Xuan W, Fan G-H. Roles of resolvins in the resolution of acute inflammation. Cell Biol Int. 2015;39:3–22. doi: 10.1002/cbin.10345. [DOI] [PubMed] [Google Scholar]
- 86.Spite M, Norling LV, Summers L, Yang R, Cooper D, et al. Resolvin D2 is a potent regulator of leukocytes and controls microbial sepsis. Nature. 2009;461:1287–91. doi: 10.1038/nature08541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Cox R, Phillips O, Fukumoto J, Fukumoto I, Tamarapu Parthasarathy P, et al. Resolvins decrease oxidative stress mediated macrophage and epithelial cell interaction through decreased cytokine secretion. PLOS ONE. 2015;10:e0136755. doi: 10.1371/journal.pone.0136755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Schmid M, Gemperle C, Rimann N, Hersberger M. Resolvin D1 polarizes primary human macrophages toward a proresolution phenotype through GPR32. J Immunol. 2016;196:3429–37. doi: 10.4049/jimmunol.1501701. [DOI] [PubMed] [Google Scholar]
- 89.Titos E, Rius B, González-Périz A, López-Vicario C, Morán-Salvador E, et al. Resolvin D1 and its precursor docosahexaenoic acid promote resolution of adipose tissue inflammation by eliciting macrophage polarization toward an M2-like phenotype. J Immunol. 2011;187:5408–18. doi: 10.4049/jimmunol.1100225. [DOI] [PubMed] [Google Scholar]
- 90.Palmer CD, Mancuso CJ, Weiss JP, Serhan CN, Guinan EC, Levy O. 17(R)-resolvin D1 differentially regulates TLR4-mediated responses of primary human macrophages to purified LPS and live E. coli. J Leukoc Biol. 2011;90:459–70. doi: 10.1189/jlb.0311145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lattin J, Zidar DA, Schroder K, Kellie S, Hume DA, Sweet MJ. G-protein-coupled receptor expression, function, and signaling in macrophages. J Leukoc Biol. 2007;82:16–32. doi: 10.1189/jlb.0107051. [DOI] [PubMed] [Google Scholar]
- 92.Brennan F, Jackson A, Chantry D, Maini R, Feldmann M. Inhibitory effect of TNFα antibodies on synovial cell interleukin-1 production in rheumatoid arthritis. Lancet. 1989;334:244–47. doi: 10.1016/s0140-6736(89)90430-3. [DOI] [PubMed] [Google Scholar]
- 93.Baldwin AG, Brough D, Freeman S. Inhibiting the inflammasome: a chemical perspective. J Med Chem. 2016;59:1691–710. doi: 10.1021/acs.jmedchem.5b01091. [DOI] [PubMed] [Google Scholar]
- 94.Rhen T, Cidlowski JA. Antiinflammatory action of glucocorticoids—new mechanisms for old drugs. N Engl J Med. 2005;353:1711–23. doi: 10.1056/NEJMra050541. [DOI] [PubMed] [Google Scholar]
- 95.McKay LI, Cidlowski JA. Molecular control of immune/inflammatory responses: interactions between nuclear factor-κB and steroid receptor-signaling pathways. Endocr Rev. 1999;20:435–59. doi: 10.1210/edrv.20.4.0375. [DOI] [PubMed] [Google Scholar]
- 96.Cato ACB, Nestl A, Mink S. Rapid actions of steroid receptors in cellular signaling pathways. Sci Signal. 2002;2002:re9. doi: 10.1126/stke.2002.138.re9. [DOI] [PubMed] [Google Scholar]
- 97.Scheinman RI, Cogswell PC, Lofquist AK, Baldwin AS. Role of transcriptional activation of IκBα in mediation of immunosuppression by glucocorticoids. Science. 1995;270:283–86. doi: 10.1126/science.270.5234.283. [DOI] [PubMed] [Google Scholar]
- 98.Erwig L-P, Henson PM. Immunological consequences of apoptotic cell phagocytosis. Am J Pathol. 2007;171:2–8. doi: 10.2353/ajpath.2007.070135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Patterson H, Nibbs R, McInnes I, Siebert S. Protein kinase inhibitors in the treatment of inflammatory and autoimmune diseases. Clin Exp Immunol. 2014;176:1–10. doi: 10.1111/cei.12248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Mócsai A, Ruland J, Tybulewicz VLJ. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat Rev Immunol. 2010;10:387–402. doi: 10.1038/nri2765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Liao C, Hsu J, Kim Y, Hu D-Q, Xu D, et al. Selective inhibition of spleen tyrosine kinase (SYK) with a novel orally bioavailable small molecule inhibitor, RO9021, impinges on various innate and adaptive immune responses: implications for SYK inhibitors in autoimmune disease therapy. Arthritis Res Ther. 2013:15R146. doi: 10.1186/ar4329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Johnson GL, Lapadat R. Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science. 2002;298:1911–12. doi: 10.1126/science.1072682. [DOI] [PubMed] [Google Scholar]
- 103.Kyriakis JM, Banerjee P, Nikolakaki E, Dai T, Rubie EA, et al. The stress-activated protein kinase subfamily of c-Jun kinases. Nature. 1994;369:156–60. doi: 10.1038/369156a0. [DOI] [PubMed] [Google Scholar]
- 104.Cuenda A, Rousseau S. P38 MAP-kinases pathway regulation, function and role in human diseases. Biochim Biophys Acta Mol Cell Res. 2007;1773:1358–75. doi: 10.1016/j.bbamcr.2007.03.010. [DOI] [PubMed] [Google Scholar]
- 105.Hommes D, Van Den Blink B, Plasse T, Bartelsman J, Xu C, et al. Inhibition of stress-activated MAP kinases induces clinical improvement in moderate to severe Crohn’s disease. Gastroenterology. 2002;122:7–14. doi: 10.1053/gast.2002.30770. [DOI] [PubMed] [Google Scholar]
- 106.Matozaki T, Murata Y, Okazawa H, Ohnishi H. Functions and molecular mechanisms of the CD47-SIRPα signalling pathway. Trends Cell Biol. 2009;19:72–80. doi: 10.1016/j.tcb.2008.12.001. [DOI] [PubMed] [Google Scholar]
- 107.Hoek RM, Ruuls SR, Murphy CA, Wright GJ, Goddard R, et al. Down-regulation of the macrophage lineage through interaction with OX2 (CD200) Science. 2000;290:1768–71. doi: 10.1126/science.290.5497.1768. [DOI] [PubMed] [Google Scholar]
- 108.Mihrshahi R, Brown MH. Downstream of tyrosine kinase 1 and 2 play opposing roles in CD200 receptor signaling. J Immunol. 2010;185:7216–22. doi: 10.4049/jimmunol.1002858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Jenmalm MC, Cherwinski H, Bowman EP, Phillips JH, Sedgwick JD. Regulation of myeloid cell function through the CD200 receptor. J Immunol. 2006;176:191–99. doi: 10.4049/jimmunol.176.1.191. [DOI] [PubMed] [Google Scholar]
- 110.Turnbull IR, Gilfillan S, Cella M, Aoshi T, Miller M, et al. Cutting edge: TREM-2 attenuates macrophage activation. J Immunol. 2006;177:3520–24. doi: 10.4049/jimmunol.177.6.3520. [DOI] [PubMed] [Google Scholar]
- 111.Daws MR, Sullam PM, Niemi EC, Chen TT, Tchao NK, Seaman WE. Pattern recognition by TREM-2: binding of anionic ligands. J Immunol. 2003;171:594–99. doi: 10.4049/jimmunol.171.2.594. [DOI] [PubMed] [Google Scholar]
- 112.Treede I, Braun A, Sparla R, Kühnel M, Giese T, et al. Anti-inflammatory effects of phosphatidylcholine. J Biol Chem. 2007;282:27155–64. doi: 10.1074/jbc.M704408200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Wang J-Y, Shieh C-C, You P-F, Lei H-Y, Reid KBM. Inhibitory effect of pulmonary surfactant proteins A and D on allergen-induced lymphocyte proliferation and histamine release in children with asthma. Am J Respir Crit Care Med. 1998;158:510–18. doi: 10.1164/ajrccm.158.2.9709111. [DOI] [PubMed] [Google Scholar]
- 114.Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O’Garra A. IL-10 inhibits cytokine production by activated macrophages. J Immunol. 1991;147:3815–22. [PubMed] [Google Scholar]
- 115.Mosser DM, Zhang X. Interleukin-10: new perspectives on an old cytokine. Immunol Rev. 2008;226:205–18. doi: 10.1111/j.1600-065X.2008.00706.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Sutterwala FS, Noel GJ, Clynes R, Mosser DM. Selective suppression of interleukin-12 induction after macrophage receptor ligation. J Exp Med. 1997;185:1977–85. doi: 10.1084/jem.185.11.1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Miles SA, Conrad SM, Alves RG, Jeronimo SMB, Mosser DM. A role for IgG immune complexes during infection with the intracellular pathogen Leishmania. J Exp Med. 2005;201:747–54. doi: 10.1084/jem.20041470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Massagué J. TGFβ signalling in context. Nat Rev Mol Cell Biol. 2012;13:616–30. doi: 10.1038/nrm3434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.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 Investig. 1998;101:890–98. doi: 10.1172/JCI1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kuruvilla AP, Shah R, Hochwald GM, Liggitt HD, Palladino MA, Thorbecke GJ. Protective effect of transforming growth factor beta 1 on experimental autoimmune diseases in mice. PNAS. 1991;88:2918–21. doi: 10.1073/pnas.88.7.2918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Racke MK, Dhib-Jalbut S, Cannella B, Albert PS, Raine CS, McFarlin DE. Prevention and treatment of chronic relapsing experimental allergic encephalomyelitis by transforming growth factor-beta 1. J Immunol. 1991;146:3012–17. [PubMed] [Google Scholar]
- 122.Willis BC, Borok Z. TGF-β-induced EMT: mechanisms and implications for fibrotic lung disease. Am J Physiol Lung Cell Mol Physiol. 2007;293:L525–34. doi: 10.1152/ajplung.00163.2007. [DOI] [PubMed] [Google Scholar]
- 123.Bhardwaj S, Srivastava N, Sudan R, Saha B. Leishmania interferes with host cell signaling to devise a survival strategy. J Biomed Biotechnol. 2010;2010:109189. doi: 10.1155/2010/109189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Hickman SP, Chan J, Salgame P. Mycobacterium tuberculosis induces differential cytokine production from dendritic cells and macrophages with divergent effects on naive T cell polarization. J Immunol. 2002;168:4636–42. doi: 10.4049/jimmunol.168.9.4636. [DOI] [PubMed] [Google Scholar]
- 125.Kim SO, Sheikh HI, Ha S-D, Martins A, Reid G. G-CSF-mediated inhibition of JNK is a key mechanism for Lactobacillus rhamnosus-induced suppression of TNF production in macrophages. Cell Microbiol. 2006;8:1958–71. doi: 10.1111/j.1462-5822.2006.00763.x. [DOI] [PubMed] [Google Scholar]
- 126.Hewitson JP, Grainger JR, Maizels RM. Helminth immunoregulation: the role of parasite secreted proteins in modulating host immunity. Mol Biochem Parasitol. 2009;167:1–11. doi: 10.1016/j.molbiopara.2009.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Gautier EL, Ivanov S, Lesnik P, Randolph GJ. Local apoptosis mediates clearance of macrophages from resolving inflammation in mice. Blood. 2013;122:2714–22. doi: 10.1182/blood-2013-01-478206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Janssen WJ, Barthel L, Muldrow A, Oberley-Deegan RE, Kearns MT, et al. Fas determines differential fates of resident and recruited macrophages during resolution of acute lung injury. Am J Respir Crit Care Med. 2011;184:547–60. doi: 10.1164/rccm.201011-1891OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Seimon T, Tabas I. Mechanisms and consequences of macrophage apoptosis in atherosclerosis. J Lipid Res. 2009;50:S382–87. doi: 10.1194/jlr.R800032-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Fox S, Leitch AE, Duffin R, Haslett C, Rossi AG. Neutrophil apoptosis: relevance to the innate immune response and inflammatory disease. J Innate Immun. 2010;2:216–27. doi: 10.1159/000284367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Huynh M-LN, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-β1 secretion and the resolution of inflammation. J Clin Investig. 2002;109:41–50. doi: 10.1172/JCI11638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Sugimoto MA, Vago JP, Teixeira MM, Sousa LP. Annexin A1 and the resolution of inflammation: modulation of neutrophil recruitment, apoptosis, and clearance. J Immunol Res. 2016;2016:8239258. doi: 10.1155/2016/8239258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Schulert GS, Grom AA. Pathogenesis of macrophage activation syndrome and potential for cytokine-directed therapies. Annu Rev Med. 2015;66:145–59. doi: 10.1146/annurev-med-061813-012806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Billiau AD, Roskams T, Van Damme-Lombaerts R, Matthys P, Wouters C. Macrophage activation syndrome: characteristic findings on liver biopsy illustrating the key role of activated, IFN-γ-producing lymphocytes and IL-6- and TNF-α-producing macrophages. Blood. 2004;105:1648–51. doi: 10.1182/blood-2004-08-2997. [DOI] [PubMed] [Google Scholar]
- 135.Behrens EM, Canna SW, Slade K, Rao S, Kreiger PA, et al. Repeated TLR9 stimulation results in macrophage activation syndrome-like disease in mice. J Clin Investig. 121:2264–77. doi: 10.1172/JCI43157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Canna SW, de Jesus AA, Gouni S, Brooks SR, Marrero B, et al. An activating NLRC4 inflamma-some mutation causes autoinflammation with recurrent macrophage activation syndrome. Nat Genet. 2014;46:1140–46. doi: 10.1038/ng.3089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Durand M, Troyanov Y, Laflamme P, Gregoire G. Macrophage activation syndrome treated with anakinra. J Rheumatol. 2010;37:879–80. doi: 10.3899/jrheum.091046. [DOI] [PubMed] [Google Scholar]
- 138.Shimizu M, Yokoyama T, Yamada K, Kaneda H, Wada H, et al. Distinct cytokine profiles of systemic-onset juvenile idiopathic arthritis-associated macrophage activation syndrome with particular emphasis on the role of interleukin-18 in its pathogenesis. Rheumatology. 2010;49:1645–53. doi: 10.1093/rheumatology/keq133. [DOI] [PubMed] [Google Scholar]
- 139.LeBouder E, Rey-Nores JE, Rushmere NK, Grigorov M, Lawn SD, et al. Soluble forms of Toll-like receptor (TLR)2 capable of modulating TLR2 signaling are present in human plasma and breast milk. J Immunol. 2003;171:6680–89. doi: 10.4049/jimmunol.171.12.6680. [DOI] [PubMed] [Google Scholar]
- 140.Ten Oever J, Kox M, van de Veerdonk FL, Mothapo KM, Slavcovici A, et al. The discriminative capacity of soluble Toll-like receptor (sTLR)2 and sTLR4 in inflammatory diseases. BMC Immunol. 2014;15:55. doi: 10.1186/s12865-014-0055-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Kitchens RL, Thompson PA, Viriyakosol S, O’Keefe GE, Munford RS. Plasma CD14 decreases monocyte responses to LPS by transferring cell-bound LPS to plasma lipoproteins. J Clin Investig. 2001;108:485–93. doi: 10.1172/JCI13139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Wu J, Hu L, Zhang G, Wu F, He T. Accuracy of presepsin in sepsis diagnosis: a systematic review and meta-analysis. PLOS ONE. 2015;10:e0133057. doi: 10.1371/journal.pone.0133057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Honda Y, Yamagiwa S, Matsuda Y, Takamura M, Ichida T, Aoyagi Y. Altered expression of TLR homolog RP105 on monocytes hypersensitive to LPS in patients with primary biliary cirrhosis. J Hepatol. 2007;47:404–11. doi: 10.1016/j.jhep.2007.03.012. [DOI] [PubMed] [Google Scholar]
- 144.Chuang TH, Ulevitch RJ. Triad3A, an E3 ubiquitin-protein ligase regulating Toll-like receptors. Nat Immunol. 2004;5:495–502. doi: 10.1038/ni1066. [DOI] [PubMed] [Google Scholar]
- 145.Chen NK, Chong TW, Loh HL, Lim KH, Gan VH, et al. Negative regulatory responses to metabolically triggered inflammation impair renal epithelial immunity in diabetes mellitus. J Mol Med. 2013;91:587–98. doi: 10.1007/s00109-012-0969-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Carty M, Goodbody R, Schroder M, Stack J, Moynagh PN, Bowie AG. The human adaptor SARM negatively regulates adaptor protein TRIF-dependent Toll-like receptor signaling. Nat Immunol. 2006;7:1074–81. doi: 10.1038/ni1382. [DOI] [PubMed] [Google Scholar]
- 147.Basith S, Manavalan B, Govindaraj RG, Choi S. In silico approach to inhibition of signaling pathways of Toll-like receptors 2 and 4 by ST2L. PLOS ONE. 2011;6:e23989. doi: 10.1371/journal.pone.0023989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Su X, Li S, Meng M, Qian W, Xie W, et al. TNF receptor-associated factor-1 (TRAF1) negatively regulates Toll/IL-1 receptor domain-containing adaptor inducing IFN-β (TRIF)-mediated signaling. Eur J Immunol. 2006;36:199–206. doi: 10.1002/eji.200535415. [DOI] [PubMed] [Google Scholar]
- 149.Liang Y, Meng S, Zhang JA, Zhu YF, Li C, et al. Tumor necrosis factor receptor-associated factor 1 (TRAF1) polymorphisms and susceptibility to autoimmune thyroid disease. Autoimmunity. 2016;49:84–89. doi: 10.3109/08916934.2015.1124423. [DOI] [PubMed] [Google Scholar]
- 150.Shen J, Qiao Y, Ran Z, Wang T. Different activation of TRAF4 and TRAF6 in inflammatory bowel disease. Mediat Inflamm. 2013;2013:647936. doi: 10.1155/2013/647936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Takeshita F, Ishii KJ, Kobiyama K, Kojima Y, Coban C, et al. TRAF4 acts as a silencer in TLR-mediated signaling through the association with TRAF6 and TRIF. Eur J Immunol. 2005;35:2477–85. doi: 10.1002/eji.200526151. [DOI] [PubMed] [Google Scholar]
- 152.Han C, Jin J, Xu S, Liu H, Li N, Cao X. Integrin CD11b negatively regulates TLR-triggered inflammatory responses by activating Syk and promoting degradation of Myd88 and TRIF via Cbl-b. Nat Immunol. 2010;11:734–42. doi: 10.1038/ni.1908. [DOI] [PubMed] [Google Scholar]
- 153.Abe T, Hirasaka K, Kohno S, Ochi A, Yamagishi N, et al. Ubiquitin ligase Cbl-b and obesity-induced insulin resistance. Endocr J. 2014;61:529–38. doi: 10.1507/endocrj.ej14-0048. [DOI] [PubMed] [Google Scholar]
- 154.Lee BC, Miyata M, Lim JH, Li JD. Deubiquitinase CYLD acts as a negative regulator for bacterium NTHi-induced inflammation by suppressing K63-linked ubiquitination of Myd88. PNAS. 2016;113:E165–71. doi: 10.1073/pnas.1518615113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Nagy N, Farkas K, Kemény L, Széll M. Phenotype-genotype correlations for clinical variants caused by CYLD mutations. Eur J Med Genet. 2015;58:271–78. doi: 10.1016/j.ejmg.2015.02.010. [DOI] [PubMed] [Google Scholar]
- 156.Ahmed CMI, Larkin J, Johnson HM. SOCS1 mimetics and antagonists: a complementary approach to positive and negative regulation of immune function. Front Immunol. 2015;6:183. doi: 10.3389/fimmu.2015.00183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Hardy MP, O’Neill LA. The murine IRAK2 gene encodes four alternatively spliced isoforms, two of which are inhibitory. J Biol Chem. 2004;279:27699–708. doi: 10.1074/jbc.M403068200. [DOI] [PubMed] [Google Scholar]
- 158.Rao N, Nguyen S, Ngo K, Fung-Leung WP. A novel splice variant of interleukin-1 receptor (IL-1R)-associated kinase 1 plays a negative regulatory role in Toll/IL-1R-induced inflammatory signaling. Mol Cell Biol. 2005;25:6521–32. doi: 10.1128/MCB.25.15.6521-6532.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Kobayashi K, Hernandez LD, Galan JE, Janeway CA, Jr, Medzhitov R, Flavell RA. IRAK-M is a negative regulator of Toll-like receptor signaling. Cell. 2002;110:191–202. doi: 10.1016/s0092-8674(02)00827-9. [DOI] [PubMed] [Google Scholar]
- 160.Lammers KM, Ouburg S, Morré SA, Crusius JBA, Gionchetti P, et al. Combined carriership of TLR9-1237C and CD14-260T alleles enhances the risk of developing chronic relapsing pouchitis. World J Gastroenterol. 2005;11:7323–29. doi: 10.3748/wjg.v11.i46.7323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.An H, Zhao W, Hou J, Zhang Y, Xie Y, et al. SHP-2 phosphatase negatively regulates the TRIF adaptor protein-dependent type I interferon and proinflammatory cytokine production. Immunity. 2006;25:919–28. doi: 10.1016/j.immuni.2006.10.014. [DOI] [PubMed] [Google Scholar]
- 162.Abu-Dayyeh I, Shio MT, Sato S, Akira S, Cousineau B, Olivier M. Leishmania-induced IRAK-1 inactivation is mediated by SHP-1 interacting with an evolutionarily conserved KTIM motif. PLOS Negl Trop Dis. 2008;2:e305. doi: 10.1371/journal.pntd.0000305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Zhu YY, Su Y, Li ZG, Zhang Y. The largely normal response to Toll-like receptor 7 and 9 stimulation and the enhanced expression of SIGIRR by B cells in systemic lupus erythematosus. PLOS ONE. 2012;7:e44131. doi: 10.1371/journal.pone.0044131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Ma A, Malynn BA. A20: linking a complex regulator of ubiquitylation to immunity and human disease. Nat Rev Immunol. 2012;12:774–85. doi: 10.1038/nri3313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Gu YJ, Sun WY, Zhang S, Wu JJ, Wei W. The emerging roles of β-arrestins in fibrotic diseases. Acta Pharmacol Sin. 2015;36:1277–87. doi: 10.1038/aps.2015.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Li H, Sun X, LeSage G, Zhang Y, Liang Z, et al. β-Arrestin 2 regulates toll-like receptor 4-mediated apoptotic signalling through glycogen synthase kinase-3β. Immunology. 2010;130:556–63. doi: 10.1111/j.1365-2567.2010.03256.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Wolska N, Rybakowska P, Rasmussen A, Brown M, Montgomery C, et al. Brief report: patients with primary Sjögren’s syndrome who are positive for autoantibodies to tripartite motif-containing protein 38 show greater disease severity. Arthritis Rheumatol. 2016;68:724–29. doi: 10.1002/art.39497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Zhao W, Wang L, Zhang M, Yuan C, Gao C. E3 ubiquitin ligase tripartite motif 38 negatively regulates TLR-mediated immune responses by proteasomal degradation of TNF receptor-associated factor 6 in macrophages. J Immunol. 2012;188:2567–74. doi: 10.4049/jimmunol.1103255. [DOI] [PubMed] [Google Scholar]
- 169.Wang Y, Shaked I, Stanford SM, Zhou W, Curtsinger JM, et al. The autoimmunity-associated gene PTPN22 potentiates toll-like receptor-driven, type 1 interferon-dependent immunity. Immunity. 2013;39:111–22. doi: 10.1016/j.immuni.2013.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Shi M, Deng W, Bi E, Mao K, Ji Y, et al. TRIM30α negatively regulates TLR-mediated NF-κB activation by targeting TAB2 and TAB3 for degradation. Nat Immunol. 2008;9:369–77. doi: 10.1038/ni1577. [DOI] [PubMed] [Google Scholar]
- 171.Gilchrist M, Thorsson V, Li B, Rust AG, Korb M, et al. Systems biology approaches identify ATF3 as a negative regulator of Toll-like receptor 4. Nature. 2006;441:173–78. doi: 10.1038/nature04768. [DOI] [PubMed] [Google Scholar]
- 172.Caso F, Galozzi P, Costa L, Sfriso P, Cantarini L, Punzi L. Autoinflammatory granulomatous diseases: from Blau syndrome and early-onset sarcoidosis to NOD2-mediated disease and Crohn’s disease. RMD Open Rheum Musculoskelet Dis. 2015;1:e000097. doi: 10.1136/rmdopen-2015-000097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Watanabe T, Kitani A, Murray PJ, Strober W. NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol. 2004;5:800–8. doi: 10.1038/ni1092. [DOI] [PubMed] [Google Scholar]
- 174.Stark A-K, Sriskantharajah S, Hessel EM, Okkenhaug K. PI3K inhibitors in inflammation, autoimmunity and cancer. Curr Opin Pharmacol. 2015;23:82–91. doi: 10.1016/j.coph.2015.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Saitoh T, Tun-Kyi A, Ryo A, Yamamoto M, Finn G, et al. Negative regulation of interferon-regulatory factor 3-dependent innate antiviral response by the prolyl isomerase Pin1. Nat Immunol. 2006;7:598–605. doi: 10.1038/ni1347. [DOI] [PubMed] [Google Scholar]
- 176.Lu Z, Hunter T. Prolyl isomerase Pin1 in cancer. Cell Res. 2014;24:1033–49. doi: 10.1038/cr.2014.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Negishi H, Ohba Y, Yanai H, Takaoka A, Honma K, et al. Negative regulation of Toll-like-receptor signaling by IRF-4. PNAS. 2005;102:15989–94. doi: 10.1073/pnas.0508327102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Manni M, Gupta S, Nixon BG, Weaver CT, Jessberger R, Pernis AB. IRF4-dependent and IRF4-independent pathways contribute to DC dysfunction in lupus. PLOS ONE. 2015;10:e0141927. doi: 10.1371/journal.pone.0141927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Ehrlich ES, Chmura JC, Smith JC, Kalu NN, Hayward GS. KSHV RTA abolishes NFκB responsive gene expression during lytic reactivation by targeting vFLIP for degradation via the proteasome. PLOS ONE. 2014;9:e91359. doi: 10.1371/journal.pone.0091359. [DOI] [PMC free article] [PubMed] [Google Scholar]