Elsevier

Clinical Immunology

Volume 173, December 2016, Pages 81-86
Clinical Immunology

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

https://doi.org/10.1016/j.clim.2016.09.005Get rights and content

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.

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 [4], [5], [6]. 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) [15], [16], [17], 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.

Section snippets

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

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

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 [58], [59], [60], [61]. 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,

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

Acknowledgements

The funding support was from the National Institute of Allergy and Infectious Diseases grant AI091526 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

References (163)

  • A. Mantovani

    The chemokine system in diverse forms of macrophage activation and polarization

    Trends Immunol.

    (2004)
  • C. Bain

    Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors

    Mucosal Immunol.

    (2013)
  • L.E. Smythies

    Inflammation anergy in human intestinal macrophages is due to Smad-induced IκBα expression and NF-κB inactivation

    J. Biol. Chem.

    (2010)
  • M. Hausmann

    Toll-like receptors 2 and 4 are up-regulated during intestinal inflammation

    Gastroenterology

    (2002)
  • M. Rescigno et al.

    Interactions among dendritic cells, macrophages, and epithelial cells in the gut: implications for immune tolerance

    Curr. Opin. Immunol.

    (2008)
  • S. Rakoff-Nahoum et al.

    Role of toll-like receptors in spontaneous commensal-dependent colitis

    Immunity

    (2006)
  • K. Takeda

    Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils

    Immunity

    (1999)
  • T. Kanai

    Interleukin 18 is a potent proliferative factor for intestinal mucosal lymphocytes in Crohn's disease

    Gastroenterology

    (2000)
  • E. Elinav

    NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis

    Cell

    (2011)
  • M. Kavai et al.

    Immune complex clearance by monocytes and macrophages in systemic lupus erythematosus

    Autoimmun. Rev.

    (2007)
  • C.G. Katsiari

    Aberrant expression of the costimulatory molecule CD40 ligand on monocytes from patients with systemic lupus erythematosus

    Clin. Immunol.

    (2002)
  • P.Y. Lee

    Type I interferon modulates monocyte recruitment and maturation in chronic inflammation

    Am. J. Pathol.

    (2009)
  • C.G. Katsiari et al.

    The pathophysiologic role of monocytes and macrophages in systemic lupus erythematosus: a reappraisal

    Semin. Arthritis Rheum.

    (2010)
  • C. Sorini et al.

    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)
  • L.W. Peterson et al.

    Intestinal epithelial cells: regulators of barrier function and immune homeostasis

    Nat. Rev. Immunol.

    (2014)
  • J. Farache

    Contributions of dendritic cells and macrophages to intestinal homeostasis and immune defense

    Immunol. Cell Biol.

    (2013)
  • T.A. Wynn et al.

    Macrophage biology in development, homeostasis and disease

    Nature

    (2013)
  • F. Ginhoux et al.

    Monocytes and macrophages: developmental pathways and tissue homeostasis

    Nat. Rev. Immunol.

    (2014)
  • F. Ginhoux

    New insights into the multidimensional concept of macrophage ontogeny, activation and function

    Nat. Immunol.

    (2016)
  • R. Xavier et al.

    Unravelling the pathogenesis of inflammatory bowel disease

    Nature

    (2007)
  • D.J. Wallace

    New insights into mechanisms of therapeutic effects of antimalarial agents in SLE

    Nat. Rev. Rheumatol.

    (2012)
  • B.J. Skaggs et al.

    Accelerated atherosclerosis in patients with SLE-mechanisms and management

    Nat. Rev. Rheumatol.

    (2012)
  • A. Mak et al.

    Global trends, potential mechanisms and early detection of organ damage in SLE

    Nat. Rev. Rheumatol.

    (2013)
  • M. Knip et al.

    The role of the intestinal microbiota in type 1 diabetes mellitus

    Nat. Rev. Endocrinol.

    (2016)
  • K.C. Herold

    Type 1 diabetes: translating mechanistic observations into effective clinical outcomes

    Nat. Rev. Immunol.

    (2013)
  • S. Viatte et al.

    Genetics and epigenetics of rheumatoid arthritis

    Nat. Rev. Rheumatol.

    (2013)
  • C.A. Dendrou et al.

    Immunopathology of multiple sclerosis

    Nat. Rev. Immunol.

    (2015)
  • F. Geissmann

    Development of monocytes, macrophages, and dendritic cells

    Science

    (2010)
  • F.K. Swirski

    Identification of splenic reservoir monocytes and their deployment to inflammatory sites

    Science

    (2009)
  • L. Ziegler-Heitbrock

    Nomenclature of monocytes and dendritic cells in blood

    Blood

    (2010)
  • L. Ziegler-Heitbrock et al.

    Toward a refined definition of monocyte subsets

    Front. Immunol.

    (2013)
  • C.C. Bain et al.

    Macrophages in intestinal homeostasis and inflammation

    Immunol. Rev.

    (2014)
  • M. Rossol

    The CD14brightCD16 + monocyte subset is expanded in rheumatoid arthritis and promotes expansion of the Th17 cell population

    Arthritis Rheum.

    (2012)
  • K.-U. Belge

    The proinflammatory CD14 + CD16 + DR ++ monocytes are a major source of TNF

    J. Immunol.

    (2002)
  • V. Hornung

    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)
  • S. Gordon et al.

    Monocyte and macrophage heterogeneity

    Nat. Rev. Immunol.

    (2005)
  • M. Guilliams

    Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny

    Nat. Rev. Immunol.

    (2014)
  • L.E. Smythies

    Mucosal IL-8 and TGF-β recruit blood monocytes: evidence for cross-talk between the lamina propria stroma and myeloid cells

    J. Leukoc. Biol.

    (2006)
  • Y. Takada

    Monocyte chemoattractant protein-1 contributes to gut homeostasis and intestinal inflammation by composition of IL-10–producing regulatory macrophage subset

    J. Immunol.

    (2010)
  • B. Weber

    CX3CR1 defines functionally distinct intestinal mononuclear phagocyte subsets which maintain their respective functions during homeostatic and inflammatory conditions

    Eur. J. Immunol.

    (2011)
  • Cited by (19)

    • A review of hematopoietic stem cell transplantation for autoimmune diseases: multiple sclerosis, systemic sclerosis and Crohn's disease. Position paper of the Brazilian Society of Bone Marrow Transplantation

      2021, Hematology, Transfusion and Cell Therapy
      Citation Excerpt :

      The mucosal disturbance may be linked to abnormalities in the toll-like recognition receptors (TLRs), which play an essential role in the pathogenesis of numerous autoimmune diseases. The perturbation of the TLR signaling pathway in intestinal macrophages has been associated with tolerance breakdown in autoimmune diseases.114 Patients with Crohn's disease present defects in the innate immune pathway, and inadequate T cell responses to pathogenic mutations in the nucleotide-binding oligomerization domain 2 (NOD2) and autophagy-related protein 16-1 (ATG16L1) genes that impair bacterial sensing and clearing.108

    • The immune reaction and degradation fate of scaffold in cartilage/bone tissue engineering

      2019, Materials Science and Engineering C
      Citation Excerpt :

      Activated M1 phenotype and the recruited macrophages can be polarized to an anti-inflammatory M2 phenotype macrophages, which is crucially related to regenerative reactions and remodeling process, by IL-4 and IL-13, and expressed high level of CD206, Ym1 (eosinophil chemotactic factor), CD163, CCL1, CCL18, FIZZ1 (Found in Inflammatory Zone protein), chitotriosidase and arginase 1. Regeneration-associated M2 macrophages can secret factors such as PDGF, VEGF, IGF1 to regulate cell mobilization, proliferation, differentiation and promote angiogenesis [153–160]. Phenotypes of macrophages can be classified by the activation standards to M(IL-4), M(IL-10), M(IFN-c) so forth as well.

    • Chasing Mavericks: The quest for defining developmental waves of hematopoiesis

      2019, Current Topics in Developmental Biology
      Citation Excerpt :

      Similarly, tissue resident macrophages (TrMacs) have been the topic of substantial interest in recent years due to their reported pre-HSC origin (Lichanska & Hume, 2000; Mizoguchi, Takahashi, Takeya, Naito, & Morioka, 1992; Sorokin, Hoyt, Blunt, & McNelly, 1992). TrMacs have been widely implicated in disease, from neurodegeneration and autoimmune disorders to cancer (Honold & Nahrendorf, 2018; Nicolás-Ávila et al., 2018; Sevenich, 2018; Yin, Valin, Dixon, & Leavenworth, 2017; Zhou, Ding, et al., 2016). Understanding how TrMacs develop and how they are maintained long-term will provide a better understanding of several diseases and may uncover potential therapeutic targets.

    • Critical Role for the Microbiota in CX <inf>3</inf> CR1 <sup>+</sup> Intestinal Mononuclear Phagocyte Regulation of Intestinal T Cell Responses

      2018, Immunity
      Citation Excerpt :

      Additionally, the microbiota is critical for induction of intestinal regulatory T (Treg) cells as they are absent in germ-free mice (Atarashi et al., 2011). Disrupted signaling downstream of the microbiota, such as through loss of toll-like receptor pathways, is thought to drive disease pathology including in IBD (Zhou et al., 2016). We and others have found that the microbiota is required for proper intestinal barrier repair through innate lymphoid cell production of IL-22 (Longman et al., 2014; Ouyang et al., 2011).

    View all citing articles on Scopus
    View full text