Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Dec 1.
Published in final edited form as: Clin Immunol. 2016 Sep 9;173:81–86. doi: 10.1016/j.clim.2016.09.005

Toll-like receptor-mediated immune responses in intestinal macrophages; implications for mucosal immunity and autoimmune diseases

Zejun Zhou 1, Miao Ding 1, Lei Huang 2, Gary Gilkeson 3, Ren Lang 4,*, Wei Jiang 1,5,*
PMCID: PMC5148676  NIHMSID: NIHMS816695  PMID: 27620642

Abstract

Monocytes are precursors of macrophages and key players during inflammation and pathogen challenge in the periphery, whereas intestinal resident macrophages act as innate effector cells to engulf and clear bacteria, secrete cytokines, and maintain intestinal immunity and homeostasis. However, perturbation of toll-like receptor signaling pathway in intestinal macrophages has been associated with tolerance breakdown in autoimmune diseases. In the present review, we have summarized and discussed the role of toll-like receptor signals in human intestinal macrophages, and the role of human intestinal macrophages in keeping human intestinal immunity, homeostasis, and autoimmune diseases.

Keywords: macrophages, monocytes, autoimmune disease, intestine

1. Introduction

The human intestinal tract harbors 1012 microorganisms per gram of luminal content, representing ten times more than that of human cells in the body [1, 2]. It is exposed constantly to massive foreign antigens and must discriminate between harmful and harmless antigens to ensure the normal function and homeostasis [3, 4]. The intestinal cells rapidly respond to different stimuli including microorganisms [46]. Meanwhile, the mucous barrier provides perfect protection against the diversity of bacteria residing in the lumen [7, 8]. Therefore, the interaction and conjunction between intestinal cells and luminal bacteria in the gut are identified to be crucial in maintaining intestinal homeostasis and are believed as “firewalls” for protection from the pathogens [1]. In the gut, mononuclear phagocyte system (MPS) maintains a delicate equilibrium between the induction of immune responses to potential pathogens and the tolerance to innocuous antigens [9]. Macrophages are major mononuclear phagocytes that play a crucial role in intestinal homeostasis and immunity [10, 11]. Monocytes in both human and mice are key players during inflammation and pathogen challenge in the periphery, whereas intestinal resident macrophages act as innate effector cells to engulf and clear bacteria or their products, secrete cytokines, and maintain intestinal homeostasis [10, 11]. However, perturbations between immunity and tolerance in the intestinal system have been shown to be associated with autoimmune diseases [12], including inflammatory bowel disease (IBD) [13, 14], systemic lupus erythematous (SLE) [1517], type 1 diabetes (T1D) [18, 19], rheumatoid arthritis (RA) [20], and multiple sclerosis (MS) [21]. In this review, we have discussed the phenotypic characterization and function of intestinal macrophages, their subpopulations, and involvement in autoimmune diseases.

2. The origin and function of monocytes and macrophages in the intestine

Monocytes are a conserved population of leukocytes and play a central role in immune system [22, 23]. The homeostasis of tissue resident macrophages relies on the constant recruitment of blood monocytes [10, 11]. Recent studies showed that resident macrophages in mice are part of the MPS that arise from the hematopoietic system, which are constituted by self-renewal hematopoietic stem cells and progenitor cells in primary lymphoid organs [10, 11]. In human, monocytes express multiple molecules, including CD14 and CD16 [24]. Consequently, monocytes are regrouped into three main subsets based on their CD14 and CD16 expression, the classical subset (CD14++CD16−), the intermediate subset (CD14++CD16+) and the non-classical subset (CD14+CD16++) [2527]. In humans, the classical monocytes are rapidly recruited to the sites of inflammation and produce IL-10 in response to the toll-like receptor (TLR) 4 ligand LPS [28]; while the classical monocytes in mice are most likely acting as the precursors of peripheral DCs and macrophages [29]. The non-classical monocytes have differential capacities to secrete key inflammatory cytokines (e.g., IL-1β, IL-6, TNF-α and sCD14) in response to TLR stimulation [30, 31]. Furthermore, monocytes are involved in innate immunity against pathogens and toxins [23, 32, 33]. Notably, monocytes in human and mice are very preponderant phagocytic cells responding to specific signals through surface molecules, including scavenger receptors (SR-A and CD36), low-density lipoprotein receptors (LRP1), TLRs (TLR1, TLR2 and TLR4), chemokine receptors (CCR2 and CX3CR1), cytokine receptors (M-CSFR), Fcγ receptors (FcγRs) and adhesion molecules (LFA-1) [33, 34]. Moreover, monocytes are important in antigen processing and presentation because of their large number in the periphery and their roles as DC progenitors [23, 35].

Recent opinions suggested that tissue macrophages in mice are derived from embryonic precursors that seed the developing tissues before birth [911]. However, a notable exception is the gastrointestinal tract, which contains large populations of resident macrophages, derived from blood monocytes in the steady state [10, 11]. A reasonable explanation is that intestinal macrophages are relatively short-lived compared to most other tissue macrophages and have very poor proliferative capacities, requiring a robust and rapid replacement from blood monocytes [28, 29, 36, 37]. Interestingly, human gut-resident macrophages do not fit into the classical “M1–M2” classification [28]. For instance, they express high levels of MHC-II and produce TNF-α constitutively that are normally associated with M1 or “classically activated” macrophages [3840]; Whereas they also express CD206, CD163, and IL-10 that are associated with M2 or M2-like macrophages [41].

Macrophages are the most abundant mononuclear phagocytes in the healthy tissues and have emerged as crucial sentinels for the maintenance of tissue homeostasis [42]. Macrophages are further defined to equip with a broad range of pathogen recognition receptors that make them efficient at phagocytosis and produce inflammatory cytokines to maintain tissue homeostasis [43]. They are characterized by surface expression of CD14, CD68, HLA-DR, as well as human epidermal growth factor module-containing mucin-like receptor 1 (EMR1) in human tissues [9, 42]. However, resident gut macrophages in murine models are identified to express high levels of CD64, Fc-g receptor 1 (FcgRI), chemokine receptors CX3CR1 and CCR2 [1, 12, 43]. In addition, macrophages in human gut are identified to closely associate with the epithelial monolayer coupled with high phagocytic and actively bactericidal, which means they are ideally located to capture and destroy any material breaching the epithelial barrier [44]. Importantly however, this is not necessary to result in the release of pro-inflammatory mediators or respiratory burst [44, 45]. Intestinal macrophages in mice also serve as antigen-presenting cells due to their high expression of MHC-II and their ability to take up antigens [12, 29, 43]. And as one of the most abundant leucocytes living in the intestinal mucosa, macrophages in mice can be activated and regulated by the prototypical Th1, Th2 cytokines, microbial or endogenous danger signals, such as IL-4, IL-13, IL-10 and TGF-β [9, 12, 46]. Recent studies have also proposed macrophage-induced IL-1β but not IL-6 in mice is critical for the development of steady-state Th17 cells, critical cells against infection, in the intestine [47, 48].

3. TLRs expression and stimulation on macrophages in gut

TLRs are key initiators of innate immune responses and promote adaptive immunity [49]. The most important cell types expressing TLRs are APCs, including macrophages, DCs, and B lymphocytes, which directly recognize various microbial pathogens through PAMPs [49, 50]. TLR engagement triggers downstream signaling pathways and ultimately results in antimicrobial responses [49, 51]. Intestinal macrophages, which represent a unique population of cells that exist in the gut, express most TLRs in humans [52]. However, hyporesponsiveness to activation via TLRs is a key feature of the resident intestinal macrophages in mice [28, 38, 47]. Intestinal macrophages contact with a biomass of bacteria, it is necessary to regulate the TLR signaling pathways because prolonged and excessive activation of TLRs can lead to uncontrolled inflammation detrimental to the host [5355]. Previous studies showed that human intestinal macrophages expressed several anti-inflammatory molecules, including IL-10, but little or no pro-inflammatory cytokines, even after stimulation with TLRs ligands [28, 44]. Intestinal macrophages have reduced CD14 surface expression and inhibit DC potential to drive Th17 T cells [56]. During the maturation from monocytes, human intestinal macrophages also down-regulate key TLR signaling molecules such as MyD88 and TRAF6, and up-regulate negative regulators such as IRAK-M and A20 [44, 52]. Moreover, intestinal macrophages in mice also continuously produce IL-10 to maintain their hyporesponsiveness to TLR ligands [12, 38]. Consistently, intestinal macrophages isolated from IL-10−/− mice robustly respond to gut bacteria, whereas wild-type intestinal macrophages are hyporesponsive [47, 48, 5759]. These data suggest the possibility that the TLR signaling pathway in macrophages in the gut may contribute to intestinal homeostasis and prevent inflammation. Interestingly, human intestinal macrophages do not produce pro-inflammatory cytokines in response to TLR ligation, but retain fully phagocytic and bactericidal activities [44].

4. The role of intestinal macrophage in autoimmune diseases

4.1 IBD

There is increasing evidence that mice knock out individual TLRs or MyD88 can trigger an abnormal inflammatory response of resident intestinal macrophages and thereby facilitate the development of IBD [5861]. As we know, to keep human intestinal homeostasis, intestinal macrophages have reduced CD14 expression and do not produce inflammatory cytokines through TLRs, although they remain phagocytic and bactericidal activities in the healthy human gut [44, 58, 62]. Moreover, mice intestinal macrophages express anti-inflammatory molecules, such as IL-10, and contribute to the differentiation of Treg cells, while suppressing DC-derived Th1 and Th17 immunities [47, 57, 63]. However, intestinal macrophages in patients with IBD robustly respond to microbial products and the resident bacteria, which results in the production of large amounts of pro-inflammatory cytokines such as TNF-α and IL-23 [64]. Consistently, colonic macrophages from mice with defective IL-10R signaling or global IL-10 cannot respond robustly to TLR stimulation, and these animals develop spontaneous colitis [6567]. On the other hand, in patients with Crohn’s disease (CD), a unique type of inflammatory macrophage population is present in the intestine, which is characterized by expressing both macrophage and DC markers (CD14, CD33, CD68, CD205, CD209), and by producing large amounts of pro-inflammatory cytokines, such as IL-23, TNF-α and IL-6 [6871]. However, these CD14-expressing lamina propria macrophages appear to contribute to IFN-γ, rather than IL-17 production, depending on IL-23 and TNF-α [72, 73].

Furthermore, administration of TLR4 ligand (LPS) or TLR9 ligand (CpG) can protect mice from colitis by increasing cyto-protective heat shock proteins or by promoting the type I IFN production, respectively [74]. In contrast, deficiencies in TLR2, TLR4, TLR5, TLR9, or MyD88 are all associated with enhanced susceptibility to colitis in mice [5, 61, 7577]. However, the precise mechanisms by which the intestinal macrophages mediate resistance against the development of colitis through TLR signaling remain unclear.

It has further been reported that the intestinal macrophages in mice also protect against colitis through TLR-independent mechanisms [78]. Besides TLRs, other PRRs expressed in intestinal macrophages such as NOD-like receptor (NLR) family of proteins are also associated with IBD [7981]. For instance, Nlrp6−/− and Nod2−/− mice have an increased susceptibility to dextran sulphate sodium (DSS)-induced colitis [82, 83]. These results suggested that the impaired recognition of microbial products by intestinal macrophages may contribute to the IBD development.

4.2 SLE

SLE is an autoimmune disease characterized by autoantibody production and chronic inflammation targeting multiple organs [1517]. Aberrant monocyte/macrophage surface markers were expressed in cells from SLE patients, including Fcγ receptors (FcγRs), ICAM-1 (intercellular adhesion molecule-1, CD54), CD40, MHC II, type-1 interferon-stimulated genes (ISGs), and sialoadhesin (Siglec-1, CD169) [8491]. In line with the deregulations of monocyte/macrophage surface markers, abnormalities in cytokine production were also exhibited in patients with SLE [9294]. Studies from ours and others found that plasma levels of sCD14, secreted by monocytes in response to LPS stimulation, are elevated, whereas the expression of membrane CD14 on monocytes are reduced in patients with SLE compared to healthy controls [95, 96]. Previous studies from our group and others showed that patients with SLE have increased monocyte number, CD16 expression, and IL-6 expression compared to healthy controls [97102], suggesting that TLR4 responses may play an important role in SLE disease pathogenesis. Notably, TLR7, TLR8 and TLR9 signaling pathways in SLE have been extensively studied [84, 95, 103, 104]. Studies in mice showed that autoantibodies against DNA and RNA-associated antigens promote the formation of nucleic acid-containing immune complexes, resulting in the activation via TLR7/8 and TLR9, thereby may be responsible for the excess production of inflammatory cytokines in patients with SLE [105, 106]. In addition, the production of two regulators of inflammatory responses (IL-10 and TNF-α) is dysregulated in SLE patients, suggesting that these cytokines may be involved in SLE disease pathogenesis [107109].

Besides the dysfunction of cytokine production, macrophages in SLE patients also have a defect in phagocytosis [110112]. Ineffective clearance of dying cells and debris by macrophages may provide a source of autoantigens for the development of autoantibodies in SLE disease [113, 114]. For instance, Denny et al. found that macrophages from lupus-prone SNF1 mice developed increases in autoantibodies against dsDNA, nucleosomes, and nephritic antibody IdLNF1 compared to untreated SNF1 mice [115]. Interestingly, a recent study showed that drinking acidic pH water promotes SNF1 mice to develop nephritis but not those on neutral pH water [116]. However, whether intestinal macrophages play a role in the induction of nephritis in response to acidic pH water is unknown.

The complement pathway was also implicated in SLE pathogenesis [117, 118]. Previous studies showed that blood monocytes express C1q receptors (cC1qR and gC1qR) as well as synthesize and secrete the classical pathway proteins C1q, C1r, C1s and C1-INH [119]. However, reduced serum levels of C1q, C3, and C4 were found in SLE, suggesting that there are maybe a direct or indirect link between complement pathway and SLE pathogenesis [119121]. It is reported that macrophages from SLE patients showed a significant defect in the internalization of apoptotic cells compared with those from healthy controls [119, 120, 122]. One reason maybe that C1q is associated with the recognition and removal of apoptotic cells; this relative C1q deficiency observed in SLE is likely to contribute to the inhibitory effect of lupus sera on the uptake of apoptotic cells by macrophages [121, 123, 124]. However, the mechanism underlying this phenomenon is largely unknown.

4.3 Type 1 diabetes

Type 1 diabetes (T1D) is an autoimmune disorder that results in the destruction of insulin-producing cells in the pancreas [3]. Aberrant PRR signaling activation through macrophages can lead to chronic inflammation and autoimmunity in the gut [125127]. On one hand, macrophages activated through a variety of PRRs signaling may play a pathogenic role in both the initiation and destruction phases of T1D [128130]. For instance, macrophages have been shown to produce IL-12 and to promote efficient differentiation of diabetogenic CD8+ cytotoxic T lymphocytes (CTLs) leading to T1D onset [131, 132]. In vitro and in vivo studies in mice and rats showed that the deleterious effect of macrophages on β-cells was mediated through the production of TNF-α and IL-1β [133, 134]. On the other hand, the dynamic adjustment of commensal bacteria in the gut may interfere with the recognition of intestinal macrophages and contribute to the development of T1D [135, 136]. For instance, pathogen-free MyD88−/− non-obese diabetic (NOD) mice that lack the ability to recognize microbial stimuli do not develop to T1D; and this effect is dependent on commensal microbes because germ-free MyD88-negative NOD mice develop to diabetes [137].

The commensal microbial community may also play an important role against T1D disease through sex hormones and preventing TLR recognition by intestinal macrophages [58, 137139]. It has been shown that the commensal microbial communities in mouse gut increased serum testosterone and protected NOD males from T1D; In contrast, transfer of gut microbiota from adult males to immature females altered the recipient’s microbiota, resulting in elevated testosterone and metabolic changes, reduced islet inflammation and autoantibody production, and protection from T1D [138]. However, much work remains to be done to understand the involvement of intestinal macrophages and sex hormones in this disease.

4.4 Rheumatoid arthritis

Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by inflammation of the synovial lining (synovitis), eventually resulting in the damage of cartilage and underlying bone [140]. The RA joint harbors a large amount of immune cell types, including monocytes, macrophages and CD4+ T cells [141]. TLR activation has been widely studied in RA [142145]. Notably, the expression of TLR2 in CD16+ blood monocytes, and that of TLR2 and TLR4 in CD14+ synovial fluid macrophages, is higher in patients with RA than those in healthy controls [146, 147]. In addition to directly promoting local inflammation by secreting pro-inflammatory mediators (TNF-α, IFNγ, IL-1β, IL-6, and IL-17), synovial monocytes/macrophages secrete chemokines that attract and maintain CD4+ T cells in the joint [145, 148]. After that, activated mouse monocytes promote CD4+ T helper cell polarization toward Th1/Th17 cells [148, 149]. Furthermore, mouse intestine macrophage-induced IL-1β is essential for Th17 cell differentiation [150], and the enhanced Th17 cell responses might be essential for progression to arthritis [151]. However, the mechanism that intestine macrophages contribute to the development of autoimmune arthritis remains unknown.

4.5 Multiple sclerosis

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) that is characterized by peripheral inflammatory processes, blood-brain barrier (BBB) breakdown, and immune cell infiltration into the CNS, leading to both axonal damage and demyelination, and ultimately resulting in disability in MS patients [152154]. Previous studies showed that high salt not only potentiates LPS-induced macrophage activation, including enhanced IL-1α, TNF-α and IL-1β production, but also enhances the development of MS partly through changing the gut microbiome [153, 155160]. Conversely, inhibition of salt-inducible kinases induces an anti-inflammatory phenotype on mice macrophages, and produces increased amount of IL-10 and decreased amount of pro-inflammatory cytokines [161]. Besides cytokines, macrophages also produce enhanced level of chemokines, defined as macrophage inflammatory protein-2 and CCL2, which are strongly depend on the salty condition [155, 162, 163]. CCL2 in particular plays an important role in the recruitment of monocytes to the CNS during experimental autoimmune encephalomyelitis (EAE) [156, 163].

5. Conclusion

To inform future research, refining the phenotypic characterization of human intestinal monocyte/macrophage subpopulations would facilitate further investigation of their involvement in autoimmune diseases. Further investigation into the direct interactions between gut microbiota and intestinal macrophages in autoimmune diseases may elucidate how macrophage responses are generated and regulated by the dynamic adjustment of microorganisms. In conclusion, an increased understanding on these fields may contribute to the understanding of autoimmune disease pathogenesis and will benefit the development of new therapeutic strategies.

Acknowledgments

The funding support was from the National Institute of Allergy and Infectious Diseases grant AI91526 and AI077283, National Institute of Arthritis and Musculoskeletal and Skin Diseases grant P60 AR062755, UL1 RR029882, the Medical Research Service at the Ralph H. Johnson VA Medical Center Merit grant VA CSRD MERIT (CX001211), BLRD MERIT (BX000470), the 12th Five Year Research Project of People’s Liberation Army (CWS11J160), Beijing Natural Science Foundation (Grant No.7152063) and Scientific Research Common Program of Beijing Municipal Commission of Education (KM2016100250017).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Zigmond E, Jung S. Intestinal macrophages: well educated exceptions from the rule. Trends Immunol. 2013;34(4):162–8. doi: 10.1016/j.it.2013.02.001. [DOI] [PubMed] [Google Scholar]
  • 2.Honda K, Takeda K. Regulatory mechanisms of immune responses to intestinal bacteria. Mucosal Immunol. 2009;2(3):187–96. doi: 10.1038/mi.2009.8. [DOI] [PubMed] [Google Scholar]
  • 3.Sorini C, Falcone M. Shaping the (auto) immune response in the gut: the role of intestinal immune regulation in the prevention of type 1 diabetes. Am J Clin Exp Immunol. 2013;2(2):156–71. [PMC free article] [PubMed] [Google Scholar]
  • 4.Schenk M, Mueller C. Adaptations of intestinal macrophages to an antigen-rich environment. Semin Immunol. 2007;2(19):84–93. doi: 10.1016/j.smim.2006.09.002. [DOI] [PubMed] [Google Scholar]
  • 5.Rakoff-Nahoum S, et al. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118(2):229–41. doi: 10.1016/j.cell.2004.07.002. [DOI] [PubMed] [Google Scholar]
  • 6.Schey R, Danzer C, Mattner J. Perturbations of mucosal homeostasis through interactions of intestinal microbes with myeloid cells. Immunobiology. 2015;220(2):227–35. doi: 10.1016/j.imbio.2014.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol. 2014;14(3):141–53. doi: 10.1038/nri3608. [DOI] [PubMed] [Google Scholar]
  • 8.Farache J, et al. Contributions of dendritic cells and macrophages to intestinal homeostasis and immune defense. Immunol Cell Biol. 2013;91(3):232–9. doi: 10.1038/icb.2012.79. [DOI] [PubMed] [Google Scholar]
  • 9.Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496(7446):445–55. doi: 10.1038/nature12034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ginhoux F, Jung S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol. 2014;14(6):392–404. doi: 10.1038/nri3671. [DOI] [PubMed] [Google Scholar]
  • 11.Ginhoux F, et al. New insights into the multidimensional concept of macrophage ontogeny, activation and function. Nat Immunol. 2016;17(1):34–40. doi: 10.1038/ni.3324. [DOI] [PubMed] [Google Scholar]
  • 12.Cerovic V, et al. Intestinal macrophages and dendritic cells: what’s the difference? Trends Immunol. 2014;35(6):270–7. doi: 10.1016/j.it.2014.04.003. [DOI] [PubMed] [Google Scholar]
  • 13.Xavier R, Podolsky D. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448(7152):427–34. doi: 10.1038/nature06005. [DOI] [PubMed] [Google Scholar]
  • 14.Geremia A, et al. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev. 2014;13(1):3–10. doi: 10.1016/j.autrev.2013.06.004. [DOI] [PubMed] [Google Scholar]
  • 15.Wallace DJ, et al. New insights into mechanisms of therapeutic effects of antimalarial agents in SLE. Nat Rev Rheumatol. 2012;8(9):522–33. doi: 10.1038/nrrheum.2012.106. [DOI] [PubMed] [Google Scholar]
  • 16.Skaggs BJ, Hahn BH, McMahon M. Accelerated atherosclerosis in patients with SLE-mechanisms and management. Nat Rev Rheumatol. 2012;8(4):214–23. doi: 10.1038/nrrheum.2012.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mak A, Isenberg DA, Lau C-S. Global trends, potential mechanisms and early detection of organ damage in SLE. Nat Rev Rheumatol. 2013;9(5):301–10. doi: 10.1038/nrrheum.2012.208. [DOI] [PubMed] [Google Scholar]
  • 18.Knip M, Siljander H. The role of the intestinal microbiota in type 1 diabetes mellitus. Nat Rev Endocrinol. 2016;12(3):154–67. doi: 10.1038/nrendo.2015.218. [DOI] [PubMed] [Google Scholar]
  • 19.Herold KC, et al. Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat Rev Immunol. 2013;13(4):243–56. doi: 10.1038/nri3422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Viatte S, Plant D, Raychaudhuri S. Genetics and epigenetics of rheumatoid arthritis. Nat Rev Rheumatol. 2013;9(3):141–53. doi: 10.1038/nrrheum.2012.237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dendrou CA, Fugger L, Friese MA. Immunopathology of multiple sclerosis. Nat Rev Immunol. 2015;15(9):545–58. doi: 10.1038/nri3871. [DOI] [PubMed] [Google Scholar]
  • 22.Xiong H, Pamer EG. Monocytes and infection: Modulator, messenger and effector. Immunobiology. 2015;220(2):210–4. doi: 10.1016/j.imbio.2014.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Geissmann F, et al. Development of monocytes, macrophages, and dendritic cells. Science. 2010;327(5966):656–61. doi: 10.1126/science.1178331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Passlick B, Flieger D, Ziegler-Heitbrock H. Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood. 1989;74(7):2527–34. [PubMed] [Google Scholar]
  • 25.Swirski FK, et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009;325(5940):612–6. doi: 10.1126/science.1175202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ziegler-Heitbrock L, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116(16):74–80. doi: 10.1182/blood-2010-02-258558. [DOI] [PubMed] [Google Scholar]
  • 27.Ziegler-Heitbrock L, Hofer T. Toward a refined definition of monocyte subsets. Front Immunol. 2013;4(23.10):3389. doi: 10.3389/fimmu.2013.00023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bain CC, Mowat AM. Macrophages in intestinal homeostasis and inflammation. Immunol Rev. 2014;260(1):102–17. doi: 10.1111/imr.12192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zigmond E, et al. Ly6C hi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells. Immunity. 2012;37(6):1076–90. doi: 10.1016/j.immuni.2012.08.026. [DOI] [PubMed] [Google Scholar]
  • 30.Rossol M, et al. The CD14brightCD16+ monocyte subset is expanded in rheumatoid arthritis and promotes expansion of the Th17 cell population. Arthritis Rheum. 2012;64(3):671–7. doi: 10.1002/art.33418. [DOI] [PubMed] [Google Scholar]
  • 31.Belge K-U, et al. The proinflammatory CD14+ CD16+ DR++ monocytes are a major source of TNF. J Immunol. 2002;168(7):3536–42. doi: 10.4049/jimmunol.168.7.3536. [DOI] [PubMed] [Google Scholar]
  • 32.Hornung V, et al. Quantitative expression of toll-like receptor 1–10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. J Immunol. 2002;168(9):4531–7. doi: 10.4049/jimmunol.168.9.4531. [DOI] [PubMed] [Google Scholar]
  • 33.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5(12):953–64. doi: 10.1038/nri1733. [DOI] [PubMed] [Google Scholar]
  • 34.Mantovani A, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677–86. doi: 10.1016/j.it.2004.09.015. [DOI] [PubMed] [Google Scholar]
  • 35.Guilliams M, et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol. 2014;14(8):571. doi: 10.1038/nri3712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Smythies LE, et al. Mucosal IL-8 and TGF-β recruit blood monocytes: evidence for cross-talk between the lamina propria stroma and myeloid cells. J Leukocyte Biol. 2006;80(3):492–9. doi: 10.1189/jlb.1005566. [DOI] [PubMed] [Google Scholar]
  • 37.Takada Y, et al. Monocyte chemoattractant protein-1 contributes to gut homeostasis and intestinal inflammation by composition of IL-10–producing regulatory macrophage subset. J Immunol. 2010;184(5):2671–6. doi: 10.4049/jimmunol.0804012. [DOI] [PubMed] [Google Scholar]
  • 38.Bain C, et al. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal immunol. 2013;6(3):498–510. doi: 10.1038/mi.2012.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Weber B, et al. CX3CR1 defines functionally distinct intestinal mononuclear phagocyte subsets which maintain their respective functions during homeostatic and inflammatory conditions. Eur J Immunol. 2011;41(3):773–9. doi: 10.1002/eji.201040965. [DOI] [PubMed] [Google Scholar]
  • 40.Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8(12):958–69. doi: 10.1038/nri2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. 2010;11(10):889–96. doi: 10.1038/ni.1937. [DOI] [PubMed] [Google Scholar]
  • 42.Davies LC, et al. Tissue-resident macrophages. Nat Immunol. 2013;14(10):986–95. doi: 10.1038/ni.2705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gautier EL, et al. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat Immunol. 2012;13(11):1118–28. doi: 10.1038/ni.2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Smythies LE, et al. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest. 2005;115(1):66–75. doi: 10.1172/JCI19229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mahida Y, Wu K, Jewell D. Respiratory burst activity of intestinal macrophages in normal and inflammatory bowel disease. Gut. 1989;30(10):1362–70. doi: 10.1136/gut.30.10.1362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Krause P, et al. IL-10-producing intestinal macrophages prevent excessive antibacterial innate immunity by limiting IL-23 synthesis. Nat Commun. 2015;6:7055. doi: 10.1038/ncomms8055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Denning TL, et al. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol. 2007;8(10):1086–94. doi: 10.1038/ni1511. [DOI] [PubMed] [Google Scholar]
  • 48.Franchi L, et al. NLRC4-driven production of IL-1β discriminates between pathogenic and commensal bacteria and promotes host intestinal defense. Nat Immunol. 2012;13(5):449–56. doi: 10.1038/ni.2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Medzhitov R. Toll-like receptors and innate immunity. Nat Rev Immunol. 2001;1(2):135–45. doi: 10.1038/35100529. [DOI] [PubMed] [Google Scholar]
  • 50.Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001;2(8):675–80. doi: 10.1038/90609. [DOI] [PubMed] [Google Scholar]
  • 51.Netea MG, Wijmenga C, O’Neill LA. Genetic variation in Toll-like receptors and disease susceptibility. Nat Immunol. 2012;13(6):535–42. doi: 10.1038/ni.2284. [DOI] [PubMed] [Google Scholar]
  • 52.Smythies LE, et al. Inflammation anergy in human intestinal macrophages is due to Smad-induced IκBα expression and NF-κB inactivation. J Biol Chem. 2010;285(25):19593–604. doi: 10.1074/jbc.M109.069955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Abreu MT, Fukata M, Arditi M. TLR signaling in the gut in health and disease. J Immunol. 2005;174(8):4453–60. doi: 10.4049/jimmunol.174.8.4453. [DOI] [PubMed] [Google Scholar]
  • 54.Hausmann M, et al. Toll-like receptors 2 and 4 are up-regulated during intestinal inflammation. Gastroenterology. 2002;122(7):1987–2000. doi: 10.1053/gast.2002.33662. [DOI] [PubMed] [Google Scholar]
  • 55.Shibolet O, Podolsky DK. TLRs in the Gut. IV. Negative regulation of Toll-like receptors and intestinal homeostasis: addition by subtraction. Am J Physiol-Gastr L. 2007;292(6):G1469–G73. doi: 10.1152/ajpgi.00531.2006. [DOI] [PubMed] [Google Scholar]
  • 56.Rescigno M, Lopatin U, Chieppa M. Interactions among dendritic cells, macrophages, and epithelial cells in the gut: implications for immune tolerance. Curr Opin Immunol. 2008;20(6):669–75. doi: 10.1016/j.coi.2008.09.007. [DOI] [PubMed] [Google Scholar]
  • 57.Kamada N, et al. Abnormally differentiated subsets of intestinal macrophage play a key role in Th1-dominant chronic colitis through excess production of IL-12 and IL-23 in response to bacteria. J Immunol. 2005;175(10):6900–8. doi: 10.4049/jimmunol.175.10.6900. [DOI] [PubMed] [Google Scholar]
  • 58.Kamada N, et al. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013;13(5):321–35. doi: 10.1038/nri3430. [DOI] [PubMed] [Google Scholar]
  • 59.Hoshi N, et al. MyD88 signalling in colonic mononuclear phagocytes drives colitis in IL-10-deficient mice. Nat Commun. 2012;3:1120. doi: 10.1038/ncomms2113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kobayashi M, et al. Toll-like receptor-dependent production of IL-12p40 causes chronic enterocolitis in myeloid cell-specific Stat3-deficient mice. J Clin Invest. 2003;111(9):1297–308. doi: 10.1172/JCI17085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Rakoff-Nahoum S, Hao L, Medzhitov R. Role of toll-like receptors in spontaneous commensal-dependent colitis. Immunity. 2006;25(2):319–29. doi: 10.1016/j.immuni.2006.06.010. [DOI] [PubMed] [Google Scholar]
  • 62.Smith PD, et al. Intestinal macrophages lack CD14 and CD89 and consequently are down-regulated for LPS-and IgA-mediated activities. J Immunol. 2001;167(5):2651–6. doi: 10.4049/jimmunol.167.5.2651. [DOI] [PubMed] [Google Scholar]
  • 63.O’Mahony C, et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-κB activation. PLoS Pathog. 2008;4(8):e1000112. doi: 10.1371/journal.ppat.1000112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kamada N, et al. Unique CD14+ intestinal macrophages contribute to the pathogenesis of Crohn disease via IL-23/IFN-γ axis. J Clin Invest. 2008;118(6):2269–80. doi: 10.1172/JCI34610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kullberg MC, et al. Helicobacter hepaticus triggers colitis in specific-pathogen-free interleukin-10 (IL-10)-deficient mice through an IL-12-and gamma interferon-dependent mechanism. Infect Immun. 1998;66(11):5157–66. doi: 10.1128/iai.66.11.5157-5166.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hirotani T, et al. The nuclear IκB protein IκBNS selectively inhibits lipopolysaccharide-induced IL-6 production in macrophages of the colonic lamina propria. J Immunol. 2005;174(6):3650–7. doi: 10.4049/jimmunol.174.6.3650. [DOI] [PubMed] [Google Scholar]
  • 67.Takeda K, et al. Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity. 1999;10(1):39–49. doi: 10.1016/s1074-7613(00)80005-9. [DOI] [PubMed] [Google Scholar]
  • 68.Kanai T, et al. Interleukin 18 is a potent proliferative factor for intestinal mucosal lymphocytes in Crohn’s disease. Gastroenterology. 2000;119(6):1514–23. doi: 10.1053/gast.2000.20260. [DOI] [PubMed] [Google Scholar]
  • 69.Fuss IJ, et al. Both IL-12p70 and IL-23 are synthesized during active Crohn’s disease and are down-regulated by treatment with anti-IL-12 p40 monoclonal antibody. Inflamm Bowel Dis. 2006;12(1):9–15. doi: 10.1097/01.mib.0000194183.92671.b6. [DOI] [PubMed] [Google Scholar]
  • 70.Grimm M, et al. Evidence for a CD14+ population of monocytes in inflammatory bowel disease mucosa-implications for pathogenesis. Clin Exp Immunol. 1995;100(2):291. doi: 10.1111/j.1365-2249.1995.tb03667.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Cua DJ, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 2003;421(6924):744–8. doi: 10.1038/nature01355. [DOI] [PubMed] [Google Scholar]
  • 72.Neurath MF. IL-23: a master regulator in Crohn disease. Nat Med. 2007;13(1):26–8. doi: 10.1038/nm0107-26. [DOI] [PubMed] [Google Scholar]
  • 73.Yen D, et al. IL-23 is essential for T cell–mediated colitis and promotes inflammation via IL-17 and IL-6. J Clin Invest. 2006;116(5):1310–6. doi: 10.1172/JCI21404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Katakura K, et al. Toll-like receptor 9–induced type I IFN protects mice from experimental colitis. J Clin Invest. 2005;115(3):695–702. doi: 10.1172/JCI22996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lowe EL, et al. Toll-like receptor 2 signaling protects mice from tumor development in a mouse model of colitis-induced cancer. PloS one. 2010;5(9):e13027. doi: 10.1371/journal.pone.0013027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Vijay-Kumar M, et al. Deletion of TLR5 results in spontaneous colitis in mice. J Clin Invest. 2007;117(12):3909–21. doi: 10.1172/JCI33084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Carvalho FA, et al. Interleukin-1β (IL-1β) promotes susceptibility of Toll-like receptor 5 (TLR5) deficient mice to colitis. Gut. 2012;61(3):373–84. doi: 10.1136/gut.2011.240556. [DOI] [PubMed] [Google Scholar]
  • 78.Chen GY, et al. The innate immune receptor Nod1 protects the intestine from inflammation-induced tumorigenesis. Cancer Res. 2008;68(24):10060–7. doi: 10.1158/0008-5472.CAN-08-2061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hugot J, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature. 2001;411(6837):599–603. doi: 10.1038/35079107. [DOI] [PubMed] [Google Scholar]
  • 80.Kobayashi KS, et al. NOD2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 2005;307(5710):731–4. doi: 10.1126/science.1104911. [DOI] [PubMed] [Google Scholar]
  • 81.Ogura Y, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature. 2001;411(6837):603–6. doi: 10.1038/35079114. [DOI] [PubMed] [Google Scholar]
  • 82.Elinav E, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145(5):745–57. doi: 10.1016/j.cell.2011.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Couturier-Maillard A, et al. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest. 2013;123(2):700–11. doi: 10.1172/JCI62236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Li Y, Lee PY, Reeves WH. Monocyte and macrophage abnormalities in systemic lupus erythematosus. Arch Immunol Ther Ex. 2010;58(5):355–64. doi: 10.1007/s00005-010-0093-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kavai M, Szegedi G. Immune complex clearance by monocytes and macrophages in systemic lupus erythematosus. Autoimmun Rev. 2007;6(7):497–502. doi: 10.1016/j.autrev.2007.01.017. [DOI] [PubMed] [Google Scholar]
  • 86.Hepburn A, Mason J, Davies K. Expression of Fcγ and complement receptors on peripheral blood monocytes in systemic lupus erythematosus and rheumatoid arthritis. Rheumatology. 2004;43(5):547–54. doi: 10.1093/rheumatology/keh112. [DOI] [PubMed] [Google Scholar]
  • 87.Funauchi M, et al. Abnormal expression of intercellular adhesion molecule-1 on peripheral blood mononuclear cells from patients with systemic lupus erythematosus. J Clin Lab Immunol. 1992;40(3):115–24. [PubMed] [Google Scholar]
  • 88.Katsiari CG, et al. Aberrant expression of the costimulatory molecule CD40 ligand on monocytes from patients with systemic lupus erythematosus. Clin Immunol. 2002;103(1):54–62. doi: 10.1006/clim.2001.5172. [DOI] [PubMed] [Google Scholar]
  • 89.Morris D, et al. MHC associations with clinical and autoantibody manifestations in European SLE. Genes Immun. 2014;15(4):210. doi: 10.1038/gene.2014.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lee PY, et al. Type I interferon modulates monocyte recruitment and maturation in chronic inflammation. Am J Pathol. 2009;175(5):2023–33. doi: 10.2353/ajpath.2009.090328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Biesen R, et al. Sialic acid-binding Ig-like lectin 1 expression in inflammatory and resident monocytes is a potential biomarker for monitoring disease activity and success of therapy in systemic lupus erythematosus. Arthritis Rheum. 2008;58(4):1136–45. doi: 10.1002/art.23404. [DOI] [PubMed] [Google Scholar]
  • 92.Katsiari CG, Liossis S-NC, Sfikakis PP. The pathophysiologic role of monocytes and macrophages in systemic lupus erythematosus: a reappraisal. Semin Arthritis Rheum. 2010;39:491–503. doi: 10.1016/j.semarthrit.2008.11.002. [DOI] [PubMed] [Google Scholar]
  • 93.Kyttaris VC, Juang Y-T, Tsokos GC. Immune cells and cytokines in systemic lupus erythematosus: an update. Curr Opin Rheumatol. 2005;17(5):518–22. doi: 10.1097/01.bor.0000170479.01451.ab. [DOI] [PubMed] [Google Scholar]
  • 94.Wong C, et al. Elevation of proinflammatory cytokine (IL-18, IL-17, IL-12) and Th2 cytokine (IL-4) concentrations in patients with systemic lupus erythematosus. Lupus. 2000;9(8):589–93. doi: 10.1191/096120300678828703. [DOI] [PubMed] [Google Scholar]
  • 95.Jiang W, et al. Sex differences in monocyte activation in systemic lupus erythematosus (SLE) PloS one. 2014;9(12):e114589. doi: 10.1371/journal.pone.0114589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Nockher W, et al. Elevated levels of soluble CD 14 in serum of patients with systemic lupus erythematosus. Clin Exp Immunol. 1994;96(1):15–9. doi: 10.1111/j.1365-2249.1994.tb06222.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Liu MF, et al. Differential expression and modulation of costimulatory molecules CD80 and CD86 on monocytes from patients with systemic lupus erythematosus. Scand J Immunol. 1999;49(1):82–7. doi: 10.1046/j.1365-3083.1999.00452.x. [DOI] [PubMed] [Google Scholar]
  • 98.Liang B, et al. Anti-interleukin-6 monoclonal antibody inhibits autoimmune responses in a murine model of systemic lupus erythematosus. Immunology. 2006;119(3):296–305. doi: 10.1111/j.1365-2567.2006.02433.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mellor-Pita S, et al. Monocytes and T lymphocytes contribute to a predominance of interleukin 6 and interleukin 10 in systemic lupus erythematosus. Cytometry B Clin Cytom. 2009;76(4):261–70. doi: 10.1002/cyto.b.20468. [DOI] [PubMed] [Google Scholar]
  • 100.Cairns AP, Crockard AD, Bell AL. The CD14+ CD16+ monocyte subset in rheumatoid arthritis and systemic lupus erythematosus. Rheumatol Int. 2002;21(5):189–92. doi: 10.1007/s00296-001-0165-8. [DOI] [PubMed] [Google Scholar]
  • 101.Yoshimoto S, et al. Elevated levels of fractalkine expression and accumulation of CD16+ monocytes in glomeruli of active lupus nephritis. Am J Kidney Dis. 2007;50(1):47–58. doi: 10.1053/j.ajkd.2007.04.012. [DOI] [PubMed] [Google Scholar]
  • 102.Nakatani K, et al. Fractalkine expression and CD16+ monocyte accumulation in glomerular lesions: association with their severity and diversity in lupus models. Am J Physiol Renal Physiol. 2010;299(1):F207–F16. doi: 10.1152/ajprenal.00482.2009. [DOI] [PubMed] [Google Scholar]
  • 103.Jiang W, Gilkeson G. Sex differences in monocytes and TLR4 associated immune responses; implications for systemic lupus erythematosus (SLE) J Immunother Appl. 2014;1:1. doi: 10.7243/2055-2394-1-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Krieg AM, Vollmer J. Toll-like receptors 7, 8, and 9: linking innate immunity to autoimmunity. Immunol Rev. 2007;220(1):251–69. doi: 10.1111/j.1600-065X.2007.00572.x. [DOI] [PubMed] [Google Scholar]
  • 105.Christensen SR, et al. Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus. Immunity. 2006;25(3):417–28. doi: 10.1016/j.immuni.2006.07.013. [DOI] [PubMed] [Google Scholar]
  • 106.Kono DH, et al. Endosomal TLR signaling is required for anti-nucleic acid and rheumatoid factor autoantibodies in lupus. Proc Natl Acad Sci USA. 2009;106(29):12061–6. doi: 10.1073/pnas.0905441106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sharif MN, et al. IFN-α priming results in a gain of proinflammatory function by IL-10: implications for systemic lupus erythematosus pathogenesis. J Immunol. 2004;172(10):6476–81. doi: 10.4049/jimmunol.172.10.6476. [DOI] [PubMed] [Google Scholar]
  • 108.Steinbach F, et al. Monocytes from systemic lupus erythematous patients are severely altered in phenotype and lineage flexibility. Ann Rheum Dis. 2000;59(4):283–8. doi: 10.1136/ard.59.4.283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Suárez A, et al. Differential effect of IL10 and TNFα genotypes on determining susceptibility to discoid and systemic lupus erythematosus. Ann Rheum Dis. 2005;64(11):1605–10. doi: 10.1136/ard.2004.035048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Janko C, et al. Inflammatory clearance of apoptotic remnants in systemic lupus erythematosus (SLE) Autoimmun Rev. 2008;8(1):9–12. doi: 10.1016/j.autrev.2008.07.015. [DOI] [PubMed] [Google Scholar]
  • 111.Gaipl US, et al. Clearance deficiency and systemic lupus erythematosus (SLE) J Autoimmun. 2007;28(2):114–21. doi: 10.1016/j.jaut.2007.02.005. [DOI] [PubMed] [Google Scholar]
  • 112.Herrmann M, et al. Impaired phagocytosis of apoptotic cell material by monocyte-derived macrophages from patients with systemic lupus erythematosus. Arthritis Rheum. 1998;41(7):1241–50. doi: 10.1002/1529-0131(199807)41:7<1241::AID-ART15>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
  • 113.Munoz L, et al. SLE-a disease of clearance deficiency? Rheumatology. 2005;44(9):1101–7. doi: 10.1093/rheumatology/keh693. [DOI] [PubMed] [Google Scholar]
  • 114.Grossmayer GE, et al. Removal of dying cells and systemic lupus erythematosus. Mod Rheumatol. 2005;15(6):383–90. doi: 10.1007/s10165-005-0430-x. [DOI] [PubMed] [Google Scholar]
  • 115.Denny MF, et al. Accelerated macrophage apoptosis induces autoantibody formation and organ damage in systemic lupus erythematosus. J Immunol. 2006;176(4):2095–104. doi: 10.4049/jimmunol.176.4.2095. [DOI] [PubMed] [Google Scholar]
  • 116.Johnson B, et al. Impact of dietary deviation on disease progression and gut microbiome composition in lupus-prone SNF1 mice. Clin Exp Immunol. 2015;181(2):323–37. doi: 10.1111/cei.12609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Leffler J, Bengtsson AA, Blom AM. The complement system in systemic lupus erythematosus: an update. Ann Rheum Dis. 2014;73(9):1601–6. doi: 10.1136/annrheumdis-2014-205287. [DOI] [PubMed] [Google Scholar]
  • 118.Elkon KB, Santer DM. Complement, interferon and lupus. Curr Opin Immunol. 2012;24(6):665–70. doi: 10.1016/j.coi.2012.08.004. [DOI] [PubMed] [Google Scholar]
  • 119.Ghebrehiwet B, et al. Monocyte expressed macromolecular C1 and C1q receptors as molecular sensors of danger: implications in SLE. Front Immunol. 2014;5:278. doi: 10.3389/fimmu.2014.00278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Manderson AP, Botto M, Walport MJ. The role of complement in the development of systemic lupus erythematosus. Annu Rev Immunol. 2004;22:431–56. doi: 10.1146/annurev.immunol.22.012703.104549. [DOI] [PubMed] [Google Scholar]
  • 121.Moosig F, et al. Reduced expression of C1q-mRNA in monocytes from patients with systemic lupus erythematosus. Clin Exp Immunol. 2006;146(3):409–16. doi: 10.1111/j.1365-2249.2006.03225.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Tas S, et al. Macrophages from patients with SLE and rheumatoid arthritis have defective adhesion in vitro, while only SLE macrophages have impaired uptake of apoptotic cells. Ann Rheum Dis. 2006;65(2):216–21. doi: 10.1136/ard.2005.037143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Benoit ME, et al. Complement protein C1q directs macrophage polarization and limits inflammasome activity during the uptake of apoptotic cells. J Immunol. 2012;188(11):5682–93. doi: 10.4049/jimmunol.1103760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Galvan MD, et al. Complement component C1q regulates macrophage expression of Mer tyrosine kinase to promote clearance of apoptotic cells. J Immunol. 2012;188(8):3716–23. doi: 10.4049/jimmunol.1102920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Biswas A, et al. Negative regulation of Toll-like receptor signaling plays an essential role in homeostasis of the intestine. Eur J Immunol. 2011;41(1):182–94. doi: 10.1002/eji.201040479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Maslowski KM, Mackay CR. Diet, gut microbiota and immune responses. Nat Immunol. 2011;12(1):5–9. doi: 10.1038/ni0111-5. [DOI] [PubMed] [Google Scholar]
  • 127.Maslowski KM, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461(7268):1282–6. doi: 10.1038/nature08530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hutchings P, et al. Transfer of diabetes in mice prevented by blockade of adhesion-promoting receptor on macrophages. Nature. 1990;348:639–42. doi: 10.1038/348639a0. [DOI] [PubMed] [Google Scholar]
  • 129.Yang L. Big mac attack: does it play a direct role for monocytes/macrophages in type 1 diabetes? Diabetes. 2008;57(11):2922–3. doi: 10.2337/db08-1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Martin AP, et al. Increased expression of CCL2 in insulin-producing cells of transgenic mice promotes mobilization of myeloid cells from the bone marrow, marked insulitis, and diabetes. Diabetes. 2008;57(11):3025–33. doi: 10.2337/db08-0625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Alleva DG, et al. Aberrant macrophage cytokine production is a conserved feature among autoimmune-prone mouse strains: elevated interleukin (IL)-12 and an imbalance in tumor necrosis factor-alpha and IL-10 define a unique cytokine profile in macrophages from young nonobese diabetic mice. Diabetes. 2000;49(7):1106–15. doi: 10.2337/diabetes.49.7.1106. [DOI] [PubMed] [Google Scholar]
  • 132.Jun H-S, et al. The role of macrophages in T cell-mediated autoimmune diabetes in nonobese diabetic mice. J Exp Med. 1999;189(2):347–58. doi: 10.1084/jem.189.2.347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Arnush M, et al. Potential role of resident islet macrophage activation in the initiation of autoimmune diabetes. J Immunol. 1998;160(6):2684–91. [PubMed] [Google Scholar]
  • 134.Dahlén E, et al. Dendritic cells and macrophages are the first and major producers of TNF-α in pancreatic islets in the nonobese diabetic mouse. J Immunol. 1998;160(7):3585–93. [PubMed] [Google Scholar]
  • 135.Lehuen A, et al. Immune cell crosstalk in type 1 diabetes. Nat Rev Immunol. 2010;10(7):501–13. doi: 10.1038/nri2787. [DOI] [PubMed] [Google Scholar]
  • 136.Burrows MP, et al. Microbiota regulates type 1 diabetes through Toll-like receptors. Proc Natl Acad Sci USA. 2015;112(32):9973–7. doi: 10.1073/pnas.1508740112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Wen L, et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature. 2008;455(7216):1109–13. doi: 10.1038/nature07336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Markle JG, et al. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science. 2013;339(6123):1084–8. doi: 10.1126/science.1233521. [DOI] [PubMed] [Google Scholar]
  • 139.Dunne J, et al. The intestinal microbiome in type 1 diabetes. Clin Exp Immunol. 2014;177(1):30–7. doi: 10.1111/cei.12321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Joosten LAB, et al. Toll-like receptors and chronic inflammation in rheumatic diseases: new developments. Nat Rev Rheumatol. 2016;12(6):344–57. doi: 10.1038/nrrheum.2016.61. [DOI] [PubMed] [Google Scholar]
  • 141.Kawashima M, et al. CD14+, CD16+ blood monocytes and joint inflammation in rheumatoid arthritis. Arthritis Rheum. 2002;46:2578–86. doi: 10.1002/art.10545. [DOI] [PubMed] [Google Scholar]
  • 142.Sokolove J, et al. Immune complexes containing citrullinated fibrinogen costimulate macrophages via Toll-like receptor 4 and Fcγ receptor. Arthritis Rheum. 2011;63(1):53–62. doi: 10.1002/art.30081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sacre SM, et al. Inhibitors of TLR8 reduce TNF production from human rheumatoid synovial membrane cultures. J Immunol. 2008;181(11):8002–9. doi: 10.4049/jimmunol.181.11.8002. [DOI] [PubMed] [Google Scholar]
  • 144.Sacre SM, et al. The Toll-like receptor adaptor proteins MyD88 and Mal/TIRAP contribute to the inflammatory and destructive processes in a human model of rheumatoid arthritis. Am J Clin Pathol. 2007;170(2):518–25. doi: 10.2353/ajpath.2007.060657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Chamberlain ND, et al. Ligation of TLR7 by rheumatoid arthritis synovial fluid single strand RNA induces transcription of TNFα in monocytes. Ann Rheum Dis. 2013;72(3):418–26. doi: 10.1136/annrheumdis-2011-201203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Iwahashi M, et al. Expression of toll-like receptor 2 on CD16+ blood monocytes and synovial tissue macrophages in rheumatoid arthritis. Arthritis Rheum. 2004;50(5):1457–67. doi: 10.1002/art.20219. [DOI] [PubMed] [Google Scholar]
  • 147.Huang Q, et al. Increased macrophage activation mediated through toll-like receptors in rheumatoid arthritis. Arthritis Rheum. 2007;56(7):2192–201. doi: 10.1002/art.22707. [DOI] [PubMed] [Google Scholar]
  • 148.Roberts CA, Dickinson AK, Taams LS. The interplay between monocytes/macrophages and CD4+ T cell subsets in rheumatoid arthritis. Front Immunol. 2015;6:571. doi: 10.3389/fimmu.2015.00571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Nakae S, et al. IL-17 production from activated T cells is required for the spontaneous development of destructive arthritis in mice deficient in IL-1 receptor antagonist. Proc Natl Acad Sci USA. 2003;100(10):5986–90. doi: 10.1073/pnas.1035999100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Wu H, et al. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity. 2010;32(6):815–27. doi: 10.1016/j.immuni.2010.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Abdollahi-Roodsaz S, et al. Stimulation of TLR2 and TLR4 differentially skews the balance of T cells in a mouse model of arthritis. J Clin Invest. 2008;118(1):205–16. doi: 10.1172/JCI32639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Yadav SK, et al. Advances in the immunopathogenesis of multiple sclerosis. Curr Opin Neurol. 2015;28(3):206–19. doi: 10.1097/WCO.0000000000000205. [DOI] [PubMed] [Google Scholar]
  • 153.Hucke S, Wiendl H, Klotz L. Implications of dietary salt intake for multiple sclerosis pathogenesis. Mult Scler. 2016;22(2):133–9. doi: 10.1177/1352458515609431. [DOI] [PubMed] [Google Scholar]
  • 154.Goldenberg MM. Multiple sclerosis review. Pharm Ther. 2012;37(3):175. [PMC free article] [PubMed] [Google Scholar]
  • 155.Ip WE, Medzhitov R. Macrophages monitor tissue osmolarity and induce inflammatory response through NLRP3 and NLRC4 inflammasome activation. Nat Commun. 2015;6:6931. doi: 10.1038/ncomms7931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Mildner A, et al. CCR2+ Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system. Brain. 2009;132:2487–500. doi: 10.1093/brain/awp144. [DOI] [PubMed] [Google Scholar]
  • 157.Jantsch J, et al. Cutaneous Na+ storage strengthens the antimicrobial barrier function of the skin and boosts macrophage-driven host defense. Cell Metab. 2015;21(3):493–501. doi: 10.1016/j.cmet.2015.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zhang W, et al. High salt primes a specific activation state of macrophages, M (Na) Cell Res. 2015;25(8):893–910. doi: 10.1038/cr.2015.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci. 2012;13(10):701–12. doi: 10.1038/nrn3346. [DOI] [PubMed] [Google Scholar]
  • 160.Joscelyn J, Kasper LH. Digesting the emerging role for the gut microbiome in central nervous system demyelination. Mult Scler. 2014;20(12):1553–9. doi: 10.1177/1352458514541579. [DOI] [PubMed] [Google Scholar]
  • 161.Clark K, et al. Phosphorylation of CRTC3 by the salt-inducible kinases controls the interconversion of classically activated and regulatory macrophages. Proc Natl Acad Sci USA. 2012;109(42):16986–91. doi: 10.1073/pnas.1215450109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Kostyk AG, et al. Regulation of chemokine expression by NaCl occurs independently of cystic fibrosis transmembrane conductance regulator in macrophages. Am J Pathol. 2006;169(1):12–20. doi: 10.2353/ajpath.2006.051042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Müller S, et al. Salt-dependent chemotaxis of macrophages. PloS one. 2013;8(9):e73439. doi: 10.1371/journal.pone.0073439. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES