Skip to main content
Virulence logoLink to Virulence
. 2013 Jan 18;4(3):207–212. doi: 10.4161/viru.23354

Toll-like receptor signaling and regulation of intestinal immunity

Karishma Kamdar 1, Vivien Nguyen 1,2, R William DePaolo 1,*
PMCID: PMC3711978  PMID: 23334153

Abstract

The intestine is a complex organ that must maintain tolerance to innocuous food antigens and commensal microbiota while being also able to mount inflammatory responses against invading pathogenic microorganisms. The ability to restrain tolerogenic responses while permitting inflammatory responses requires communication between commensal bacteria, intestinal epithelial cells and immune cells. Disruption or improper signaling between any of these factors may lead to uncontrolled inflammation and the development of inflammatory diseases. Toll-like receptors (TLR) recognize conserved molecular motifs of microorganisms and, not surprisingly, are important for maintaining tolerance to commensal microbiota, as well as inducing inflammation against pathogens. Perturbations in individual TLR signaling can lead to a number of different outcomes and illustrate a system of regulation within the intestine in which each TLR plays a largely non-redundant role in mucosal immunity. This review will discuss recent findings on the roles of individual TLRs and intestinal homeostasis.

Keywords: TLR, intestine, mucosal immunity, commensal microbiota, inflammation, tolerance

Introduction

Our intestine contains 100 trillion commensal bacteria,1 outnumbering our human cells by a ratio of 10:1. Surprisingly, our intestine is able to maintain tolerance to this incredible antigenic burden, yet still provide protective inflammatory responses against invading enteric pathogens. Homeostasis refers to the dynamic balance in the intestine between tolerance and inflammation suggesting the existence of specific mechanisms that are able to restrain inflammation, during steady-state conditions. Perturbations of these mechanisms may result in dysregulation of intestinal responses leading to dysbiosis of commensal populations, aberrant immune responses and ultimately development of inflammatory disease.

One can consider that the gastrointestinal tract contains three distinct compartments. The microbial component encompasses luminal or mucosal-associated commensal bacteria and, in humans is composed of 6–10 phyla and approximately 5,000 distinct species.2 Changes in the microbiota, termed dysbiosis, have been associated with chronic intestinal diseases such as inflammatory bowel disease (IBD),3 as well as with extraintestinal diseases such as diabetes4 or multiple sclerosis.5,6 The immune compartment is contained within the lamina propria and the Peyer’s patches. In these sites naïve T cells are induced to become regulatory T cells or pro-inflammatory effector cells via specialized dendritic cells (DC).7 Separating the immune cells and the commensal microbes is a single layer of cells comprising the intestinal epithelium. The intestinal epithelium is made up of a number of cell types with functions that range from cytokine secretion, IgA secretion, production of anti-microbial peptides and mucus production.

Due to the diverse functions and complicated nature of these intestinal compartments the regulation and maintenance of homeostasis is important to prevent dysregulated immune responses and development of inflammatory disease. Given the intimate association between the microbial world and the gut, it is not surprising that innate signals are critical for maintenance of intestinal homeostasis. TLRs comprise a set of innate molecules that recognize conserved molecular motifs on bacteria and viruses and are present on both epithelial and immune cells.8 Activation of a TLR by its ligand induces several intracellular signaling cascades resulting in the production of cytokines, chemokines and the transcription of other genes important for initiating and controlling infection. The myeloid differentiation primary response gene (MyD88) adaptor molecule is involved in many signaling pathways including TLR signaling (excluding TLR3) and non-TLR pathways such as IL-1R signaling pathway.9 Studies using mice deficient for MyD88 have shown that signaling via TLRs plays an important role in intestinal homeostasis. Recognition of commensal microbiota in a MyD88-dependent manner has been shown to be required for epithelial cell homeostasis,10 response to injury,11 and induction of antimicrobial peptides.12,13

Since MyD88 is a common signaling molecule for most TLRs, one may reason that individual effects of a single TLR would either be represented in the phenotype of MyD88-deficient mice or would be redundant among all the TLRs. However, the reported effects of individual TLRs on mucosal homeostasis seem to point to distinct and non-redundant roles. This review will highlight the impact of those TLRs that recognize bacteria, fungi, and parasites on intestinal homeostasis and disease.

TLR2 and Its Co-Receptors, TLR1 and TLR6

TLR2 recognizes molecular patterns associated with Gram-negative and Gram-positive bacteria and yeast, i.e., lipoproteins,14,15 lipoteichoic acid16 and zymosan,17 respectively. Ligand-induced activation of TLR2 leads to recruitment of toll-interleukin receptor domain containing adaptor protein (TIRAP) and MyD88, which results in activation of nuclear factor kappa b (NFκB), and production of cytokines and chemokines.18,19 TLR2 is functionally expressed by a number of distinct cell types in the intestinal mucosa and is constitutively expressed in the murine gastrointestinal epithelium, but expression is restricted to the crypts.20 In the healthy gut, intestinal epithelial cells restrain potential pro-inflammatory responses to ubiquitous commensal lipoproteins through modulation of TLR2 signaling.21 These alterations in TLR2 signaling occur as a result of increased expression of negative regulators, such as Tollip and A20,22 and activation of cell signaling pathways23 that induce production of anti-inflammatory IL-10, which inhibits macrophage and DC effector functions and limits immune responses.24 Alterations in TLR2-mediated NFκB activation in antigen-presenting cells(APC), as seen in CARD15 (NOD2) mutations, leads to mucosal inflammation through exaggerated IFN-γ, IL-12 and IL-23 production.25

During acute intestinal injury or inflammation, stimulation of TLR2/1 heterodimers by Pam3CysSK4, a synthetic triacylated lipoprotein analog, preserves zonula occludens-1-associated barrier via activation of PI3K and Akt.10,24 However, in chronic intestinal inflammation, such as that induced by adoptive transfer of naïve CD4+ T cells into mice lacking adaptive immunity (RAG1-deficient mice), TLR2 signaling does not affect gut pathology.26 Therefore, TLR2 signaling may confer protection against acute mechanical injury through maintenance of tight junction integrity while having minimal effects on regulation of sustained inflammatory processes.

Apart from its role in barrier defense and immune responses in the mucosal compartment, TLR2 signaling has direct and indirect effects on T cell function. TLR2 signaling in APC induces expression of enzymes involved in the metabolism of vitamin A and allows the APC to imprint a gut-homing phenotype on T cells.27 Direct activation of TLR2/1 on human CD4+ T cells promotes TH17 responses,28 and TLR2/1 signaling on committed CD4+ TReg cells reduces their suppressive activity by promoting a shift toward IL-17 production.29 However, polysaccharide A (PSA), a product of the commensal bacteria B. fragilis, binds TLR2 independent of TLR1 or TLR6 and promotes IL-10 production in CD4+ T cells, thereby restraining TH17 responses and enhancing its own colonization of the gut,30 suggesting that commensal bacteria are able to exploit TLR pathways to suppress immunity, thereby establishing host-microbial symbiosis. These seemingly contradictory effects are likely due to differences in co-receptor engagement and highlight the multiple and opposing functions attributed to TLR2.

The ability of TLR2 signaling to produce pro- and anti-inflammatory responses may be due to its ability to interact with multiple co-receptors,31 including TLR1,32 TLR6,32 Dectin-1,33 CD3634 and CD14.35 However, this characteristic, which sets it apart from other TLRs, makes it difficult to study.36 Our group has shown that TLR2/6 ligands educate DC to become tolerogenic and promote the polarization of IL-10-producing regulatory T cells (Tr1) in vitro and in vivo.37 On the other hand, activation of TLR2/1 educates DC to produce greater amounts of IL12p40 and low levels of IL-10, promoting the differentiation of TH137 or TH17 cells.31 These findings demonstrate that engagement of a specific co-receptor by TLR2 can promote either an inflammatory or a regulatory response. Alteration in the inflammatory outcome is due to the difference in activation of cell signaling pathways. Release of IL12p40 by TLR2/1-activated DC is caused by P38-MAPK activation, whereas the regulatory response by TLR2/6 DC is dependent on JNK activation.37

It is becoming increasingly evident that not only microbial signals but also the tissue microenvironment contribute to the type of immune response generated. Interestingly, our work and the work of others show an important role for the generation of mucosal immune responses via TLR2/1 signaling. Infection or elevated levels of IL-6 have been shown to help induce mucosal IL-17 responses in conjunction with mucosal production of TGF-β.38,39 We have identified that specific TLR2/1 signaling in mucosal DC during oral infection with Yersinia enterocolitica induces an increase in IL-6 and IL-23 and prime TH17 responses that clear the infection.31 On the other hand, TLR2/1 signaling in the spleen during intravenous infection induces IL-12p70 and primes TH1 immunity.31 Within the gut, the epithelial cells express TLR121 and TLR1 signaling in the epithelial cells indirectly impacts the generation of TH17 priming via the production of chemokines, which recruit DC to the site of infection (manuscript in review). Also unique to the gut tissue is the production of IgA, and TLR1 signaling is critical for the generation of Yersinia-specific secretory IgA in the feces of mice.31 Interestingly, TLR1 signaling in DC outside of the gut can imprint a gut-homing phenotype on T cells via the induction of enzymes that promote the metabolism of vitamin A.27

TLR2/6 signaling leads to the induction of IL-10 and immunosuppression via a secreted virulence factor of Yersinia species31,37 and the synthetic diacylated ligand FSL-1.37 Unlike TLR2/1 signaling, TLR2/6 induction of IL-10 is ubiquitous, found both in mucosal31 and systemic tissues,31,37 suggesting that TLR6 may be important for regulating or dampening immune responses. Overall, these studies emphasize that interaction of TLR2 with either TLR1 or TLR6 leads to dual and opposing effects. Given that TLR2/1 and TLR2/6 ligands are triacylated40 and diacylated41 lipoproteins, respectively, this suggests that the bacteria can modulate the immune response and can evade host immunity depending upon its acylation status.42

TLR2 activation by commensal bacteria has been shown to be involved in extraintestinal diseases. For example in experimental encephalomyelitis (EAE), a mouse model of multiple sclerosis, a lipid derived from Porphyromonas gingivalis and other bacteria commonly found in the gastrointestinal tract are capable of enhancing autoimmunity in a TLR2-dependent manner.43 However, the role of TLR2 in direct regulation of the enteric microbiota has not yet been examined; therefore it is unclear whether the shifts in microbiota influence extra-intestinal diseases. Even less described is the interaction of TLR1 or TLR6 with TLR2 that could affect outcomes of extraintestinal diseases.

TLR4

TLR4 has been shown to be involved in defense against pathogens as well as establishing commensal colonization and maintaining tolerance to commensal bacteria. However, despite a common signaling pathway that includes recruitment of MyD88, phosphorylation of IL1-receptor-associated kinase (IRAK) and tumor necrosis factor receptor-associated factor 6 (TRAF6), and subsequent release of NFκB and IFNβ,44,45 the downstream effects of TLR4 are varied. This range of responses may depend on the inflammatory status of the mucosal microenvironment.

Though barely detectable at baseline, TLR4 expression and sensitivity to its ligand, lipopolysaccharide (LPS), are increased in the setting of intestinal injury associated with Crohn disease and ulcerative colitis,46,47 and in the presence of inflammatory cytokines such as IFN-γ and TNF-α.48 Upon disruption of the epithelium, activation of TLR4 elicits inflammatory cytokine and chemokine expression with recruitment of innate and adaptive immune cells to limit bacterial invasion. For instance, in enteric Toxoplasma gondii infection and in dextran-sodium sulfate (DSS) colitis, TLR4 signaling leads to increased IL-6 and IL-12 expression49 and neutrophil recruitment,50 respectively, which contain bacterial translocation. Similarly, B cell recruitment and IgA production via induction of CCL20, CCL28 and proliferation-inducing ligand (APRIL) occur during constitutive activation of TLR445 to assist in clearance of pathogens.51 The absence of TLR4 signaling during injury results in a pattern of severe mucosal damage with impaired epithelial proliferation, attenuated inflammatory response and marked bacterial translocation.50 Though maladaptive in the short-term, the restrained proliferation associated with aberrant TLR4 signaling confers protection from malignancy associated with chronic colonic inflammation.44,52

TLR4 signaling has also been shown to affect the intestinal flora. Regulation of the microbiota by TLR4 appears to be attributable to alterations in gastrointestinal motility, which may assist in clearance of pathogens and maintenance of commensal populations,53 differentiation of goblet cells,54 and expression of antimicrobial peptides. TLR4 is constitutively expressed in the mouse gastrointestinal crypts20 where its signaling directly regulates transcription of α-defensin genes13 and β-defensin-2 genes55 in response to alterations in the microbial communities of the gut. TLR4 signaling is also involved in the development of antibodies to biliary epithelial cells in primary sclerosing cholangitis, a disease that is closely associated with IBD and is thought to arise from impaired intestinal mucosal integrity. This finding demonstrates that defective TLR4 signaling may cause a shift in intestinal microbiota and the development of extraintestinal diseases.56

TLR5

TLR5 recognizes flagellin, the main protein of bacterial flagella and is crucial for the detection of invasive flagellated bacteria at the mucosal surface.57 TLR5 plays an important role in maintaining intestinal homeostasis by regulating host defense against enterobacterial infections. This is in part because of the differential expression pattern of TLR5. Mucosal but not splenic DC express TLR5 due to different host environments including the presence of retinoic acid and other host stromal factors that alter TLR5 expression.58 Activation of TLR5 signaling induces mucosal production of IL-17 and IL-22, which promote antimicrobial defense important for clearance of the pathogen. Production of TLR5-dependent IL-17, and IL-22 can occur via DC-dependent activation of CD3CD127+ lymphoid tissue inducer cells (LTi),59,60 activation of TH1/TH17 cells, decreased expression of TReg cells60 and decreased induction mucosal IgA production.61

The recognition of flagellin by TLR5 is the dominant means by which model intestinal epithelia activate pro-inflammatory gene expression in response to Salmonella enterica. However, TLR5 knockout (TLR5KO) mice are resistant to Salmonella, and this resistance is attributed to changes in the basal phenotype of TLR5KO mice.62 The small intestine and colon of TLR5KO mice exhibit elevated levels of host defense genes that mediate innate and adaptive immunity in the gut. This includes changes in the basal phenotype of antimicrobial peptides and an increase in serum and fecal IgA and IgG and transport proteins in the gut.63 TLR5KO mice also have a homeostatic shift in microbiota composition with an increase in Proteobacteria, more specifically enterobacterial species including E. coli, which was observed in proximity to the gut epithelium.64 Whether the change in microbiota composition (due to absence of TLR5 signaling) or the absence of TLR5 directly contributes to the change in basal phenotype is not clearly understood.

The absence of TLR5 signaling leads to increased resistance to infection, dysbiosis, alterations in gene expression and also impacts host metabolism. Naïve TLR5KO mice exhibit the hallmark features of metabolic syndrome including increases in body mass, visceral fat, triglycerides, cholesterol, blood pressure and low-grade chronic inflammation. TLR5KO mice also have insulin resistance even when on a calorie-restricted diet. Loss of TLR5-signaling in RAG1-deficient mice, which lack T and B lymphocytes, still results in impaired glucose regulation, demonstrating that development of TLR5KO metabolic syndrome occurs independently of the adaptive immune system. Transfer of TLR5KO microbiota to wild-type germ-free mice conferred many aspects of the TLR5KO phenotype, suggesting that the altered microbiota contributes to the development of metabolic syndrome.65 However, whether the altered microbiota is the cause or the effect in TLR5KO mice remains yet to be determined.

TLR9

TLR9 is localized intracellularly in the endosomal compartment and recognizes intracellular bacteria by binding unmethylated cytosine phosphate guanine (CpG) dinucleotides.66 These nucleotides are expressed at high levels in prokaryotic DNA found within the commensal microbiome. Studies examining the localization of TLR9 in intestinal epithelial cells have suggested that activation can occur via basolateral and apical surface domains of TLR9.67 However, the possibility of an endosomal response in epithelial cells cannot be overruled, as differential sorting of the endosomes may occur.68 These studies suggest that the signaling of TLR9 on the apical or basolateral surfaces determine whether the response is tolerogenic or inflammatory, respectively. Apical activation of TLR9 does not induce NFκB. However, it induces expression of Frizzled 5, a regulator or Paneth cell maturation, and is for the production of antimicrobial peptides.67 In contrast, basolateral activation of TLR9 activates NFκB activation and ultimately induces IL-8 production.67 Regulating tolerance and inflammation upon the surface of the epithelial cells makes sense as the apical surface faces the intestinal lumen and comes into contact with commensal bacteria, and probiotic DNA suppresses inflammation and is protective in models of colitis.69,70 Furthermore the maintenance of Paneth cells and their secreted antimicrobial peptides appears to be important for homeostatic control of the commensal bacteria. In contrast to commensal bacteria, pathogenic bacteria that have breached the epithelium would stimulate basolateral TLR9 to produce inflammatory mediators and initiate the immune response.

During inflammation the gut must control the differentiation of TReg cells, which have the potential to limit the inflammatory response. In the absence of TLR9 there is an increase in Treg cells within the small intestine, leading to an inability to protect from infection.71 The administration of CpG to antibiotic treated mice either infected with Toxoplasma gondii71 or during chemical-induced colitis72 acts as an adjuvant and contributes to the inflammatory response.

Conclusion

Altogether, these data demonstrate that TLR signaling in the intestine has important and non-redundant effects on regulating the commensal microbiota, inducing inflammatory responses and restraining or promoting tolerance. Globally, pathogens contain ligands for multiple TLR, and in any infection or inflammatory state multiple signaling pathways will be activated and will influence each other. As discussed in this review, TLR expression can be compartmentalized to specific cell types and locations in the gut, and individual TLR may induce a different set of cytokines (e.g., TLR6 and the production of IL-10). Thus there are many levels of regulation that allow specific TLR engagements to fine-tune the immune response. This fine-tuning of the immune response by TLR ligands can be manipulated by commensal microbiota in order to allow colonization, promote tolerance and limit inflammatory disease. In turn, manipulation of TLR signaling by pathogens represents an important evasion strategy. Understanding how unique TLR signals can impact different cellular responses is important for the generation of potential oral vaccines or in the treatment of inflammatory disease.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Footnotes

References

  • 1.Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr. 2002;22:283–307. doi: 10.1146/annurev.nutr.22.011602.092259. [DOI] [PubMed] [Google Scholar]
  • 2.Carvalho FA, Aitken JD, Vijay-Kumar M, Gewirtz AT. Toll-like receptor-gut microbiota interactions: perturb at your own risk! Annu Rev Physiol. 2012;74:177–98. doi: 10.1146/annurev-physiol-020911-153330. [DOI] [PubMed] [Google Scholar]
  • 3.Sun L, Nava GM, Stappenbeck TS. Host genetic susceptibility, dysbiosis, and viral triggers in inflammatory bowel disease. Curr Opin Gastroenterol. 2011;27:321–7. doi: 10.1097/MOG.0b013e32834661b4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stonebraker AC, et al. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature. 2008;455:1109–13. doi: 10.1038/nature07336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4615–22. doi: 10.1073/pnas.1000082107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ochoa-Repáraz J, Mielcarz DW, Ditrio LE, Burroughs AR, Foureau DM, Haque-Begum S, et al. Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis. J Immunol. 2009;183:6041–50. doi: 10.4049/jimmunol.0900747. [DOI] [PubMed] [Google Scholar]
  • 7.Rescigno M. Functional specialization of antigen presenting cells in the gastrointestinal tract. Curr Opin Immunol. 2010;22:131–6. doi: 10.1016/j.coi.2009.12.007. [DOI] [PubMed] [Google Scholar]
  • 8.Santaolalla R, Fukata M, Abreu MT. Innate immunity in the small intestine. Curr Opin Gastroenterol. 2011;27:125–31. doi: 10.1097/MOG.0b013e3283438dea. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Casanova JL, Abel L, Quintana-Murci L. Human TLRs and IL-1Rs in host defense: natural insights from evolutionary, epidemiological, and clinical genetics. Annu Rev Immunol. 2011;29:447–91. doi: 10.1146/annurev-immunol-030409-101335. [DOI] [PubMed] [Google Scholar]
  • 10.Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229–41. doi: 10.1016/j.cell.2004.07.002. [DOI] [PubMed] [Google Scholar]
  • 11.Malvin NP, Seno H, Stappenbeck TS. Colonic epithelial response to injury requires Myd88 signaling in myeloid cells. Mucosal Immunol. 2012;5:194–206. doi: 10.1038/mi.2011.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Vaishnava S, Behrendt CL, Ismail AS, Eckmann L, Hooper LV. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc Natl Acad Sci U S A. 2008;105:20858–63. doi: 10.1073/pnas.0808723105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Menendez A, Willing BP, Montero M, Wlodarska M, So CC, Bhinder G, et al. Bacterial Stimulation of the TLR-MyD88 Pathway Modulates the Homeostatic Expression of Ileal Paneth Cell α-Defensins. J Innate Immun. 2013;5:39–49. doi: 10.1159/000341630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Brightbill HD, Libraty DH, Krutzik SR, Yang RB, Belisle JT, Bleharski JR, et al. Host defense mechanisms triggered by microbial lipoproteins through toll-like receptors. Science. 1999;285:732–6. doi: 10.1126/science.285.5428.732. [DOI] [PubMed] [Google Scholar]
  • 15.Aliprantis AO, Yang RB, Mark MR, Suggett S, Devaux B, Radolf JD, et al. Cell activation and apoptosis by bacterial lipoproteins through toll-like receptor-2. Science. 1999;285:736–9. doi: 10.1126/science.285.5428.736. [DOI] [PubMed] [Google Scholar]
  • 16.Schwandner R, Dziarski R, Wesche H, Rothe M, Kirschning CJ. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J Biol Chem. 1999;274:17406–9. doi: 10.1074/jbc.274.25.17406. [DOI] [PubMed] [Google Scholar]
  • 17.Underhill DM, Ozinsky A, Hajjar AM, Stevens A, Wilson CB, Bassetti M, et al. The Toll-like receptor 2 is recruited to macrophage phagosomes and discriminates between pathogens. Nature. 1999;401:811–5. doi: 10.1038/44605. [DOI] [PubMed] [Google Scholar]
  • 18.Horng T, Barton GM, Flavell RA, Medzhitov R. The adaptor molecule TIRAP provides signalling specificity for Toll-like receptors. Nature. 2002;420:329–33. doi: 10.1038/nature01180. [DOI] [PubMed] [Google Scholar]
  • 19.Yamamoto M, Sato S, Hemmi H, Sanjo H, Uematsu S, Kaisho T, et al. Essential role for TIRAP in activation of the signalling cascade shared by TLR2 and TLR4. Nature. 2002;420:324–9. doi: 10.1038/nature01182. [DOI] [PubMed] [Google Scholar]
  • 20.Ortega-Cava CF, Ishihara S, Rumi MA, Kawashima K, Ishimura N, Kazumori H, et al. Strategic compartmentalization of Toll-like receptor 4 in the mouse gut. J Immunol. 2003;170:3977–85. doi: 10.4049/jimmunol.170.8.3977. [DOI] [PubMed] [Google Scholar]
  • 21.Melmed G, Thomas LS, Lee N, Tesfay SY, Lukasek K, Michelsen KS, et al. Human intestinal epithelial cells are broadly unresponsive to Toll-like receptor 2-dependent bacterial ligands: implications for host-microbial interactions in the gut. J Immunol. 2003;170:1406–15. doi: 10.4049/jimmunol.170.3.1406. [DOI] [PubMed] [Google Scholar]
  • 22.Boone DL, Turer EE, Lee EG, Ahmad RC, Wheeler MT, Tsui C, 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]
  • 23.Cario E, Gerken G, Podolsky DK. Toll-like receptor 2 controls mucosal inflammation by regulating epithelial barrier function. Gastroenterology. 2007;132:1359–74. doi: 10.1053/j.gastro.2007.02.056. [DOI] [PubMed] [Google Scholar]
  • 24.Cario E. Barrier-protective function of intestinal epithelial Toll-like receptor 2. Mucosal Immunol. 2008;1(Suppl 1):S62–6. doi: 10.1038/mi.2008.47. [DOI] [PubMed] [Google Scholar]
  • 25.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]
  • 26.Mowat AM. Does TLR2 regulate intestinal inflammation? Eur J Immunol. 2010;40:318–20. doi: 10.1002/eji.200940232. [DOI] [PubMed] [Google Scholar]
  • 27.Wang S, Villablanca EJ, De Calisto J, Gomes DC, Nguyen DD, Mizoguchi E, et al. MyD88-dependent TLR1/2 signals educate dendritic cells with gut-specific imprinting properties. J Immunol. 2011;187:141–50. doi: 10.4049/jimmunol.1003740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Reynolds JM, Pappu BP, Peng J, Martinez GJ, Zhang Y, Chung Y, et al. Toll-like receptor 2 signaling in CD4(+) T lymphocytes promotes T helper 17 responses and regulates the pathogenesis of autoimmune disease. Immunity. 2010;32:692–702. doi: 10.1016/j.immuni.2010.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nyirenda MH, Sanvito L, Darlington PJ, O’Brien K, Zhang GX, Constantinescu CS, et al. TLR2 stimulation drives human naive and effector regulatory T cells into a Th17-like phenotype with reduced suppressive function. J Immunol. 2011;187:2278–90. doi: 10.4049/jimmunol.1003715. [DOI] [PubMed] [Google Scholar]
  • 30.Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, et al. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science. 2011;332:974–7. doi: 10.1126/science.1206095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.DePaolo RW, Kamdar K, Khakpour S, Sugiura Y, Wang W, Jabri B. A specific role for TLR1 in protective T(H)17 immunity during mucosal infection. J Exp Med. 2012;209:1437–44. doi: 10.1084/jem.20112339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ozinsky A, Underhill DM, Fontenot JD, Hajjar AM, Smith KD, Wilson CB, et al. The repertoire for pattern recognition of pathogens by the innate immune system is defined by cooperation between toll-like receptors. Proc Natl Acad Sci U S A. 2000;97:13766–71. doi: 10.1073/pnas.250476497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gantner BN, Simmons RM, Canavera SJ, Akira S, Underhill DM. Collaborative induction of inflammatory responses by dectin-1 and Toll-like receptor 2. J Exp Med. 2003;197:1107–17. doi: 10.1084/jem.20021787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K, et al. CD36 is a sensor of diacylglycerides. Nature. 2005;433:523–7. doi: 10.1038/nature03253. [DOI] [PubMed] [Google Scholar]
  • 35.Hirschfeld M, Kirschning CJ, Schwandner R, Wesche H, Weis JH, Wooten RM, et al. Cutting edge: inflammatory signaling by Borrelia burgdorferi lipoproteins is mediated by toll-like receptor 2. J Immunol. 1999;163:2382–6. [PubMed] [Google Scholar]
  • 36.Cleveland MG, Gorham JD, Murphy TL, Tuomanen E, Murphy KM. Lipoteichoic acid preparations of gram-positive bacteria induce interleukin-12 through a CD14-dependent pathway. Infect Immun. 1996;64:1906–12. doi: 10.1128/iai.64.6.1906-1912.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Depaolo RW, Tang F, Kim I, Han M, Levin N, Ciletti N, et al. Toll-like receptor 6 drives differentiation of tolerogenic dendritic cells and contributes to LcrV-mediated plague pathogenesis. Cell Host Microbe. 2008;4:350–61. doi: 10.1016/j.chom.2008.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–8. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
  • 39.Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M, et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007;317:256–60. doi: 10.1126/science.1145697. [DOI] [PubMed] [Google Scholar]
  • 40.Jin MS, Kim SE, Heo JY, Lee ME, Kim HM, Paik SG, et al. Crystal structure of the TLR1-TLR2 heterodimer induced by binding of a tri-acylated lipopeptide. Cell. 2007;130:1071–82. doi: 10.1016/j.cell.2007.09.008. [DOI] [PubMed] [Google Scholar]
  • 41.Kang JY, Nan X, Jin MS, Youn SJ, Ryu YH, Mah S, et al. Recognition of lipopeptide patterns by Toll-like receptor 2-Toll-like receptor 6 heterodimer. Immunity. 2009;31:873–84. doi: 10.1016/j.immuni.2009.09.018. [DOI] [PubMed] [Google Scholar]
  • 42.Omueti KO, Beyer JM, Johnson CM, Lyle EA, Tapping RI. Domain exchange between human toll-like receptors 1 and 6 reveals a region required for lipopeptide discrimination. J Biol Chem. 2005;280:36616–25. doi: 10.1074/jbc.M504320200. [DOI] [PubMed] [Google Scholar]
  • 43.Nichols FC, Housley WJ, O’Conor CA, Manning T, Wu S, Clark RB. Unique lipids from a common human bacterium represent a new class of Toll-like receptor 2 ligands capable of enhancing autoimmunity. Am J Pathol. 2009;175:2430–8. doi: 10.2353/ajpath.2009.090544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Fukata M, Hernandez Y, Conduah D, Cohen J, Chen A, Breglio K, et al. Innate immune signaling by Toll-like receptor-4 (TLR4) shapes the inflammatory microenvironment in colitis-associated tumors. Inflamm Bowel Dis. 2009;15:997–1006. doi: 10.1002/ibd.20880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Shang L, Fukata M, Thirunarayanan N, Martin AP, Arnaboldi P, Maussang D, et al. Toll-like receptor signaling in small intestinal epithelium promotes B-cell recruitment and IgA production in lamina propria. Gastroenterology. 2008;135:529–38. doi: 10.1053/j.gastro.2008.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cario E, Podolsky DK. Differential alteration in intestinal epithelial cell expression of toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect Immun. 2000;68:7010–7. doi: 10.1128/IAI.68.12.7010-7017.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Abreu MT, Vora P, Faure E, Thomas LS, Arnold ET, Arditi M. Decreased expression of Toll-like receptor-4 and MD-2 correlates with intestinal epithelial cell protection against dysregulated proinflammatory gene expression in response to bacterial lipopolysaccharide. J Immunol. 2001;167:1609–16. doi: 10.4049/jimmunol.167.3.1609. [DOI] [PubMed] [Google Scholar]
  • 48.Suzuki M, Hisamatsu T, Podolsky DK. Gamma interferon augments the intracellular pathway for lipopolysaccharide (LPS) recognition in human intestinal epithelial cells through coordinated up-regulation of LPS uptake and expression of the intracellular Toll-like receptor 4-MD-2 complex. Infect Immun. 2003;71:3503–11. doi: 10.1128/IAI.71.6.3503-3511.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Furuta T, Kikuchi T, Akira S, Watanabe N, Yoshikawa Y. Roles of the small intestine for induction of toll-like receptor 4-mediated innate resistance in naturally acquired murine toxoplasmosis. Int Immunol. 2006;18:1655–62. doi: 10.1093/intimm/dxl099. [DOI] [PubMed] [Google Scholar]
  • 50.Fukata M, Michelsen KS, Eri R, Thomas LS, Hu B, Lukasek K, et al. Toll-like receptor-4 is required for intestinal response to epithelial injury and limiting bacterial translocation in a murine model of acute colitis. Am J Physiol Gastrointest Liver Physiol. 2005;288:G1055–65. doi: 10.1152/ajpgi.00328.2004. [DOI] [PubMed] [Google Scholar]
  • 51.Wijburg OL, Uren TK, Simpfendorfer K, Johansen FE, Brandtzaeg P, Strugnell RA. Innate secretory antibodies protect against natural Salmonella typhimurium infection. J Exp Med. 2006;203:21–6. doi: 10.1084/jem.20052093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Fukata M, Shang L, Santaolalla R, Sotolongo J, Pastorini C, España C, et al. Constitutive activation of epithelial TLR4 augments inflammatory responses to mucosal injury and drives colitis-associated tumorigenesis. Inflamm Bowel Dis. 2011;17:1464–73. doi: 10.1002/ibd.21527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Anitha M, Vijay-Kumar M, Sitaraman SV, Gewirtz AT, Srinivasan S. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling. Gastroenterology. 2012;143:1006–16, e4. doi: 10.1053/j.gastro.2012.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sodhi CP, Neal MD, Siggers R, Sho S, Ma C, Branca MF, et al. Intestinal epithelial Toll-like receptor 4 regulates goblet cell development and is required for necrotizing enterocolitis in mice. Gastroenterology. 2012;143:708–18, e1-5. doi: 10.1053/j.gastro.2012.05.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Vora P, Youdim A, Thomas LS, Fukata M, Tesfay SY, Lukasek K, et al. Beta-defensin-2 expression is regulated by TLR signaling in intestinal epithelial cells. J Immunol. 2004;173:5398–405. doi: 10.4049/jimmunol.173.9.5398. [DOI] [PubMed] [Google Scholar]
  • 56.Karrar A, Broomé U, Södergren T, Jaksch M, Bergquist A, Björnstedt M, et al. Biliary epithelial cell antibodies link adaptive and innate immune responses in primary sclerosing cholangitis. Gastroenterology. 2007;132:1504–14. doi: 10.1053/j.gastro.2007.01.039. [DOI] [PubMed] [Google Scholar]
  • 57.Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett DR, et al. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature. 2001;410:1099–103. doi: 10.1038/35074106. [DOI] [PubMed] [Google Scholar]
  • 58.Feng T, Cong Y, Qin H, Benveniste EN, Elson CO. Generation of mucosal dendritic cells from bone marrow reveals a critical role of retinoic acid. J Immunol. 2010;185:5915–25. doi: 10.4049/jimmunol.1001233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Van Maele L, Carnoy C, Cayet D, Songhet P, Dumoutier L, Ferrero I, et al. TLR5 signaling stimulates the innate production of IL-17 and IL-22 by CD3(neg)CD127+ immune cells in spleen and mucosa. J Immunol. 2010;185:1177–85. doi: 10.4049/jimmunol.1000115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Smith KD, Andersen-Nissen E, Hayashi F, Strobe K, Bergman MA, Barrett SL, et al. Toll-like receptor 5 recognizes a conserved site on flagellin required for protofilament formation and bacterial motility. Nat Immunol. 2003;4:1247–53. doi: 10.1038/ni1011. [DOI] [PubMed] [Google Scholar]
  • 61.Cong Y, Feng T, Fujihashi K, Schoeb TR, Elson CO. A dominant, coordinated T regulatory cell-IgA response to the intestinal microbiota. Proc Natl Acad Sci U S A. 2009;106:19256–61. doi: 10.1073/pnas.0812681106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Uematsu S, Jang MH, Chevrier N, Guo Z, Kumagai Y, Yamamoto M, et al. Detection of pathogenic intestinal bacteria by Toll-like receptor 5 on intestinal CD11c+ lamina propria cells. Nat Immunol. 2006;7:868–74. doi: 10.1038/ni1362. [DOI] [PubMed] [Google Scholar]
  • 63.Vijay-Kumar M, Aitken JD, Kumar A, Neish AS, Uematsu S, Akira S, et al. Toll-like receptor 5-deficient mice have dysregulated intestinal gene expression and nonspecific resistance to Salmonella-induced typhoid-like disease. Infect Immun. 2008;76:1276–81. doi: 10.1128/IAI.01491-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Carvalho FA, Koren O, Goodrich JK, Johansson ME, Nalbantoglu I, Aitken JD, et al. Transient inability to manage proteobacteria promotes chronic gut inflammation in TLR5-deficient mice. Cell Host Microbe. 2012;12:139–52. doi: 10.1016/j.chom.2012.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, Mwangi S, Srinivasan S, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science. 2010;328:228–31. doi: 10.1126/science.1179721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ishii KJ, Akira S. Innate immune recognition of, and regulation by, DNA. Trends Immunol. 2006;27:525–32. doi: 10.1016/j.it.2006.09.002. [DOI] [PubMed] [Google Scholar]
  • 67.Lee J, Mo JH, Katakura K, Alkalay I, Rucker AN, Liu YT, et al. Maintenance of colonic homeostasis by distinctive apical TLR9 signalling in intestinal epithelial cells. Nat Cell Biol. 2006;8:1327–36. doi: 10.1038/ncb1500. [DOI] [PubMed] [Google Scholar]
  • 68.Van IJzendoorn SC, Maier O, Van Der Wouden JM, Hoekstra D. The subapical compartment and its role in intracellular trafficking and cell polarity. J Cell Physiol. 2000;184:151–60. doi: 10.1002/1097-4652(200008)184:2<151::AID-JCP2>3.0.CO;2-R. [DOI] [PubMed] [Google Scholar]
  • 69.Rachmilewitz D, Katakura K, Karmeli F, Hayashi T, Reinus C, Rudensky B, et al. Toll-like receptor 9 signaling mediates the anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology. 2004;126:520–8. doi: 10.1053/j.gastro.2003.11.019. [DOI] [PubMed] [Google Scholar]
  • 70.Rachmilewitz D, Karmeli F, Takabayashi K, Hayashi T, Leider-Trejo L, Lee J, et al. Immunostimulatory DNA ameliorates experimental and spontaneous murine colitis. Gastroenterology. 2002;122:1428–41. doi: 10.1053/gast.2002.32994. [DOI] [PubMed] [Google Scholar]
  • 71.Hall JA, Bouladoux N, Sun CM, Wohlfert EA, Blank RB, Zhu Q, et al. Commensal DNA limits regulatory T cell conversion and is a natural adjuvant of intestinal immune responses. Immunity. 2008;29:637–49. doi: 10.1016/j.immuni.2008.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Obermeier F, Dunger N, Strauch UG, Hofmann C, Bleich A, Grunwald N, et al. CpG motifs of bacterial DNA essentially contribute to the perpetuation of chronic intestinal inflammation. Gastroenterology. 2005;129:913–27. doi: 10.1053/j.gastro.2005.06.061. [DOI] [PubMed] [Google Scholar]

Articles from Virulence are provided here courtesy of Taylor & Francis

RESOURCES