Abstract
Regulatory T cells play a crucial role in normal gut homeostasis, as well as during infection with microbial or parasitic pathogens. Prior to infection, interactions with the commensal microflora are essential to differentiation of a healthy steady-state level of immunoregulation, mediated through both Toll-like receptor-dependent and -independent pathways. The ingress of pathogenic organisms may, according to the context, promote or reverse the regulatory environment, with onward consequences for inflammation in both the intestinal and extra-intestinal settings. Appropriate regulation of gut immunity thus depends upon a complex three-way interplay between host cells, commensals and pathogens, and can exert a major impact on systemic responses including allergy and autoimmunity.
Keywords: Colitis, helminths, inflammatory bowel disease, microbiome, protozoa
Introduction
In the gastrointestinal (GI) tract, the immune system is faced with the most demanding of all decision-making, with little room for error. It is imperative at all times to discriminate between, and respond correctly to, beneficial symbionts, harmless food antigens and potential pathogens [1]. There is increasing appreciation that regulatory T cells (Tregs) play a prominent and essential role in maintaining appropriate responsiveness in the gut [2,3], actively enforcing homeostasis and preventing untoward immune responses occurring. While stimulated by specific antigens, of both self and non-self origin, Tregs can transcend antigen specificity, mediating bystander suppression in a manner likely to modify systemic immune status as suggested by the ‘hygiene hypothesis’.
The steady-state: commensals, no infection
Recent studies have changed our perspective of commensal microbes from benign but inert passengers to active participants in both the postnatal development of mucosal immunity and in its long-term steady-state function. Germ-free mice show extensive deficiencies in intestinal immune system development, with reduced lymphoid tissue and fewer lymphocytes [4]. The CD4+ T cell population is diminished, affecting T helper type 1 (Th1) cells disproportionately although, remarkably, Treg frequencies are maintained or increased in germ-free mice. These and other data have established that defined components of the gut flora can play a major role in intestinal homeostasis, including protection against gut injury and mediating oral tolerance against dietary antigens.
In mice which acquire a conventional microbiome, the immune system develops normally while maintaining a continuing dialogue with the commensal population. Here, one of the dominant roles of Tregs is to prevent exuberant responses against gut flora, with which the intestinal tract is in intimate contact. Nevertheless, how commensals communicate with cells to ensure immune homeostasis is still unclear. One critical factor in this interaction at the molecular level is the host Toll-like receptor (TLR) system, as demonstrated by spontaneous colitis in TLR-5-deficient mice [5]. Where colitis is induced experimentally (e.g. by dextran sulphate administration), the absence of TLR signalling then results in greatly aggravated pathology, again indicating that TLR-mediated recognition of commensal molecules contributes to dampening immune reactivity [6]. The requirement for TLR signalling in induction of oral tolerance to dietary antigens [7] also speaks to the bimodal participation of the TLR system in both stimulatory and regulatory arms of the immune response.
Recent evidence suggests that TLR signalling can impact Treg homeostasis and that Tregs themselves express TLRs selectively. Hence, interaction with certain ligands, such as those binding TLR-2, can favour Treg expansion both in vitro and in vivo[8], while TLR-9-mediated recognition of DNA from gut flora is an essential step in ensuring that effector T cells are able, when appropriate, to overcome Treg inhibition and mount an immune response [9].
Alteration of the structural integrity of TLR signalling components is often associated with profound clinical outcome and susceptibility to various infections or autoimmune disorders. During conditions of floral translocation, peripheral TLR-9 signalling is a crucial mediator of polymicrobial sepsis. Moreover, in other conditions in which bacterial translocation occurs [for example, during irradiation and human immunodefiency virus (HIV) infection] peripheral TLR-4 signals enhance the activation status of both CD4+ and CD8+ T cells [10]. However, under most circumstances the tissues of the GI tract are exposed constantly to TLR ligands harboured by the commensal gut flora.
Mice deficient in TLR-9 display increased frequencies of Tregs within intestinal effector sites and reduced levels of constitutive interleukin (IL)-17- and interferon (IFN)-γ-producing effector T cells [9]. Complementing this, lamina propria dendritic cells (DCs) lacking exposure to gut flora DNA, induce Treg conversion in vitro. Furthermore, Tregversus effector T cell disequilibrium in TLR-9−/− mice restricts immune responses to oral infection with the pathogen Encephalitozoon cuniculi. Impaired intestinal immune responses were recapitulated in mice treated with antibiotics and were reversible after reconstitution with gut flora DNA [9]. Thus, signals derived from the gut flora act as adjuvants of immune responses for priming intestinal responses against oral pathogens via modulation of the equilibrium between Treg and effector T cells.
Intestinal epithelial cell (IEC) expression of TLRs has also proved to be important in maintaining the homeostatic host–microbiome relationship, and to involve unexpected subtleties. For example, TLR-9 is expressed on both the apical (luminal-facing) and basolateral surfaces of the epithelial cell layer, but only basolateral ligation triggers an inflammatory signal, while apical binding is inhibitory [11]. The capacity of IECs to control immune responsiveness extends to the production of thymic stromal lymphopoietin (TSLP) and IL-25, influencing the Th phenotype balance in a manner which can make or break effective immunity [12].
Not all commensals are equal
The structure and composition of the gut flora reflect natural selection at both the microbial and host levels, and show perturbations in GI dysfunction. For example, modified gut floral composition is found in inflammatory bowel disease (IBD) patients [13]. Furthermore, the presence of certain bacteria can aggravate small intestinal immunopathology following oral infection. There can be exquisite specificity in the effects of individual commensal species; for example, within the genus Lactobacillus different species are associated with differing in vivo allergic status in infants [14] and contrasting capacity to induce regulatory cytokines in vitro[15]. Moreover, even different strains or mutants of particular Lactobacillus species stimulated very different immunological outcomes in mice [16,17].
Recent evidence demonstrates that colonization of germ-free mice with complex microbiota orchestrated a broad spectrum of Th1, Th17 and Treg responses. Whereas most tested individual bacteria failed to stimulate intestinal T cell responses efficiently, a restricted number of individual bacteria can control the tonicity of the gut immune system [18]. The key commensal organisms in immune system development have been identified very recently as segmented filamentous bacteria [18,19]. A further reflection of how the make-up of the intestinal flora can impact upon systemic responses is found in studies of non-obese diabetic (NOD) mice, which succumb spontaneously to type 1 diabetes (T1D); it has been known for some time that higher microbial exposure militates against development of this autoimmune disease [20], but it was shown recently not only that conventionally housed myeloid differentiation primary response gene 88 (MyD88)−/− mice are resistant to T1D, but that resistance to disease is due to the distinct microbial combination with which they are colonized. Hence, MyD88−/− mice develop T1D under germ-free conditions, while wild-type mice given the microbial population from MyD88−/− animals had reduced susceptibility to disease [21].
It is tempting to speculate that alteration of Treg homeostasis mediated by TLR signalling, either because of genetic polymorphism or because of changes in gut flora composition, could also have consequences on development of gut inflammatory disorders. Indeed, gut flora bacteria are not equal in their capacity to stimulate TLR-9 and do so with various levels of efficiency that correlate with the frequency of cytosine–guanine dinucleotides. Thus, control of the Treg ratio and effector T cell function in the GI tract is likely to be regulated differentially by specific gut flora species. An illustration of how the presence of defined bacterial species can influence the outcome of an infection comes from the observation that mice fed Bifidobacterium infantis are protected from the pathogenic effect and translocation of Salmonella[22]. Activation of Tregs by the probiotic microorganism contributed to this protective effect.
Can commensals regulate pathology?
The proposition that certain commensal species may act in a counterinflammatory manner has led to extensive investigation of potential probiotic regulation of immunopathology. Promising results have been obtained with probiotics in the treatment of human inflammatory diseases of the intestine and in the prevention and treatment of atopic eczema in neonates and infants, but mechanism(s) of action remain to be elucidated [23]. In mice, probiotic Lactobacillus and/or Bifidobacterium treatment suppressed trinitrobenzene sulphonic acid (TNBS)-induced colitis [24,25], as well as allergy, while raising transforming growth factor (TGF)-β production [26] and stimulating Tregs able to transfer down-modulation of allergy [27]. There is also good evidence of probiotic modulation of DCs towards a proregulatory function [15,28]. Of course, not all commensals are down-regulatory, and some (like Helicobacter hepaticus) may be pathogenic in some settings, yet induce Tregs in others [29]. Furthermore, there can be significant interactions between pathogens, as in the example of intestinal bacteria aggravating the immunopathology caused by Toxoplasma infection [30]. In the latter setting, there is reduced floral complexity, either because of relative loss of more ‘regulatory’ strains or simply as a broad reflection of an altered homeostasis accompanying pathogenesis.
One consequence of the immune system's reliance on microflora for optimal immunoregulation is that antibiotic therapies may result in unintended activation of immune effector mechanisms. In model systems, antibiotic treatment renders mice more susceptible to induction of food allergy [7] as well as allergic airway inflammation [31]. For the human population, antibiotics are seen as major modifiers of beneficial human–microbe interactions [32] superimposed upon alterations caused by other exogenous factors including urbanization, global travel and dietary changes [33]. The acute effects of antibiotic treatment on the native gut microbiota range from self-limiting diarrhoea to life-threatening pseudomembranous colitis induced by bacteria filling the niche provided by the reduction in bacterial diversity [34]. The long-term consequences of such perturbations for the human–microbial symbiosis are more difficult to discern, but chronic conditions such as asthma and atopic disease have been associated with childhood antibiotic use and an altered intestinal microbiota [35–37].
Because many chemical transformations in the gut are mediated by specific microbial populations, with implications for, among others, cancer and obesity, changes in the composition of the gut microbiota could have important but undiscovered health effects. In this regard, ciprofloxacin treatment of healthy volunteers influenced the abundance of about a third of the bacterial taxa in the gut, decreasing the taxonomic richness, diversity and evenness of the community. However, the magnitude of this effect varied among individuals, and some taxa showed interindividual variation in the response to ciprofloxacin. In each individual, the taxonomic composition of the community closely resembled its pretreatment state by 4 weeks after the end of treatment, but several taxa failed to recover within 6 months [38].
The production of active anti-inflammatory mediators by particular commensal species (reviewed in [39]) provides a mechanistic framework for microbial regulation of pathology in the GI tract. One of the most striking examples is the zwitterionic polysaccharide A of Bacterioides fragilis, which restores CD4+ T cell numbers in germ-free mice [40] and protects mice against Helicobacter-induced colitis with the induction of IL-10-producing Tr1 cells [41]. Hence, immunoregulation may revolve around highly specific host–microbial molecular interactions, presumably reflecting a long and intimate co-evolution of the symbiotic relationship.
De novo induction of Tregs in the GI tract
The vitamin A metabolite, retinoic acid (RA), plays a major role in the GI tract, via its capacity to enhance the TGF-β-mediated generation of forkhead box P3 (FoxP3+) Tregs from naive T cells by gut DCs [42]. Reciprocally, RA can inhibit the generation of Th17 cells [43], suggesting that it may play an important role in maintaining the balance between effector and regulatory populations in the GI tract. Several populations of mucosal APC can induce Tregs via RA, although only the CD103 subset is equipped with the enzymatic machinery to generate RA.
Retinoic acid can also imprint gut homing molecules on various populations of lymphocytes. Defined microenvironments may have evolved self-contained strategies in which local mediators (such as RA) can imprint homing properties while also favouring the induction or function of Tregs. It is therefore tempting to speculate that a link between homing and regulatory function induction may represent a more general mechanism. Such a strategy could allow the constant generation and migration of Tregs to defined compartments. These Tregs would be expected to have the prerequisite antigen specificities (e.g. persistent microorganisms, flora antigens), status of activation and survival requirement that allow them to regulate a defined microenvironment.
Although the capacity of gut-associated lymphoid tissue (GALT) DCs or macrophages to imprint gut-homing receptors and induce FoxP3+ Tregs is associated with their capacity to release RA, it remains unclear if these cells are the main producers of this metabolite in the gut. Synthesis of RA from stored or dietary retinol depends on the direct expression of the appropriate enzymes by GALT DCs. Certainly, DCs from Peyer's patches and mesenteric lymph nodes (MLNs) express Aldh1a1 and Aldh1a2, respectively, and CD103+ DCs from the lamina propria express a large array of this family of enzymes; moreover, Peyer's patch and MLN DCs can convert retinol directly to RA in culture. However, other cells, including IELs, can express enzymes associated with vitamin A metabolism, suggesting that DCs may also acquire retinoic acid from other sources and store it. A recent study demonstrated that monocyte-derived DCs pretreated with RA can acquire several attributes characteristic of mucosal DCs, such as secretion of TGF-β and IL-6, and the capacity to augment mucosal homing receptor expression and IgA responses in lymphocytes [44]. In this particular study, these gut-derived features acquired by DCs were associated with the capacity of DCs to become carriers and not producers of RA. The precise factors that govern the activation of some of these enzymes as well as how inflammation or infections modify the metabolism of vitamin A remain to be explored. Importantly, how commensals contribute to the expression of these enzymes and metabolism of vitamin A remains unknown. Another important question is the timing necessary for DCs migrating in the GALT to acquire RA from epithelial cells and how these processes can be modified during infection. How RA contributes to oral tolerance, and at the same time protective immunity in the GI tract, also remains to be addressed. One possibility is that RA favours the induction of Tregs in the absence of secondary signals but enhances effector responses following exposure to inflammatory mediators.
Treg populations require not only appropriate conditions for their induction, but also for their upkeep, particularly when confronted with an inflammatory environment. Very recently it has been shown that, in the gut, myeloid cell-derived IL-10 plays a crucial role in maintaining functional Treg activity by stimulating IL-10R directly on FoxP3+ Tregs and allowing them to play a fully protective role in the prevention of colitis [45]. Thus, in the absence of either innate IL-10 production, or IL-10R on Tregs, these cells lose the ability to block colitogenic effector T cells from causing inflammatory disease, and indeed succumb themselves to the inflammatory process by switching to the production of IFN-γ[45]. Hence, IL-10 is important for the maintenance of Treg activity and can be pivotal at the tipping-point between regulation and inflammation.
Systemic effects of GI regulation
The regulation of Treg activity between the gut and the periphery is also of special interest, as IBD in humans may affect extraintestinal organs in up to 36% of cases [46]. IBD-related extraintestinal disorders are not specific to IBD. They can be classified into reactive manifestations dependent directly upon intestinal disease. The often co-existing presentation in the same patient points towards common underlying pathomechanisms that may involve enteric flora activating the immune system to turn against bacterial antigens and, based on cross-reactivity, against intestinal antigens and antigens in extraintestinal organs (‘molecular mimicry’).
A separate subset of IBD patients shows an increased frequency of other common autoimmune diseases that manifest mainly independently of the bowel disease. This may thus reflect susceptibility to autoimmunity in general.
The complex relationship between intestinal and extraintestinal manifestations in IBD is also reflected by the complex multi-genetic control reported in animal models of IBD; genetic loci regulating intestinal and extra-intestinal manifestations are largely but not exclusively different [47].
Parasite infection: pro- or counter-regulatory?
The appearance of GI parasites is a major challenge to the discriminatory powers of the immune system, and one which in evolutionary time has been played out countless times. The host rarely fails to respond to the infection, but the outcome in terms of response mode (Th1, Th2, Th17) and the degree of Treg activation varies markedly according the pathogen in question. In recent years, good experimental data has been provided to show that host regulatory pathways are activated by certain GI parasites in particular helminths. For example, the duodenal-dwelling nematode Heligmosomoides polygyrus can inhibit gut inflammation in the mouse associated with Helicobacter colitis [48], genetic IL-10 deficiency [49] or peanut allergy [50]; the same parasite stimulates Treg expansion and induction in vivo and in vitro[51–53]. In Trichuris muris infections of the colon, Tregs are required to minimize intestinal pathology and the parasite strain able to survive longest in the mouse is associated with the largest numerical expansion in Tregs[54].
Although data from human helminth infections are not so definitive, new and remarkable evidence has been provided for the presence of GI helminth-associated Tregs. A cohort of multiple sclerosis patients were found to have acquired gut helminth infections while under longitudinal monitoring in the clinic; infected individuals showed a dramatically lower rate of relapse, with milder clinical scores, than case–controlled uninfected patients. Infected subjects showed higher correlates of Treg activity and lower inflammatory cytokine production on autoantigen stimulation, linking the helminth infection with expanded Treg activity and improved clinical outcome [55].
Studies to date have not been defined whether the Treg subsets stimulated by GI helminths are natural or induced, or if there are parasite-specific Treg populations among them. In addition, the relative importance of Tr1 (non-FoxP3-expressing, IL-10-producing) regulatory cells is brought into question by the dispensible nature of IL-10 for many helminth-associated regulatory effects (for example [56]). By contrast, new data are clearly demonstrating an inherent capacity to promote induced Treg development and function in the case of H. polygyrus secretions which drive de novo expression of FoxP3 in naive peripheral T cells.
The distinction between Tregs and inducible regulatory T cells in vivo is not always clear, particularly in highly inflammatory settings. Moreover, Tregs may be able to influence the emergence or function of one another. This notion was suggested recently in a model of Aspergillus conidia infection in mice. In this model, control of allergic immunopathology induced by the fungus required the sequential activity of various populations of Tregs[57]. This sequential role for various populations of Tregs may not be an exception but rather the rule, as most infections proceed through various stages and therefore require various layers of regulation.
The host, on the other hand, has many mechanisms which may uphold or restore responsiveness in a counter-regulatory fashion. For example, upon infectious challenge the gut-resident APC are likely to be replaced by inflammatory cells that have not been conditioned by the gut environment [58]. Previous reports examining both gut and lung inflammation support the idea that restricted or defective Treg conversion can enhance immunopathology [59]. Such limitations of conversion during inflammation raise the possibility that exposure to antigen at a time of acute infection may impair the acquisition of tolerance against commensals that could, in turn, contribute further to the pathological process. Whatever the mix of factors at play, it is clear that regulation by pathogens is a dynamic process and, under the right circumstances, host immunity can reassert itself to overcome the infection.
Systemic consequences and long-term effects of GI infections
If changes in the commensal population within the GI tract impact upon systemic immune responses, as discussed above, then it is not surprising to find that parasitic infections in the same milieu can also exert substantial systemic effects. The influence of infection on ‘bystander’ responses, particularly where mediated through various regulatory cell populations, provides a mechanistic explanation of the more general ‘hygiene hypothesis’ concept that increasing rates of allergy and asthma in western countries could be the consequence of reduced infectious stresses during early childhood [60].
Experimental work has lent strong support for this hypothesis. For example, during GI infection, helminth-driven Treg suppression of effector function protects against subsequent airway inflammation [56]. Similar infections change responses to blood-stage malaria [61] and interfere with vaccinations [62,63]. Evidence for bystander suppression in human GI helminth infection is also accumulating, with lower allergy rates in infected children [64,65], and lower inflammatory responses to autoantigen in the multiple sclerosis study mentioned above [55]. Indeed, helminth therapy is being trialled as a potential strategy to ameliorate intestinal inflammation in Crohn's disease and ulcerative colitis [66]. Notably, other suppressive cell types are observed in these infections, including ‘regulatory B cells’ and alternatively activated macrophages, although the interdependence and sequence of activation of these other regulatory components have yet to be discerned [67].
Pathogens may therefore have evolved to exploit, and even imitate, our symbiotic relationship with gut flora. As described above, probiotic microorganisms have beneficial effects in the treatment of inflammatory bowel diseases through the induction of Treg populations, and evidence is now emerging that some helminths can act similarly. As with commensal microbes, different helminths exert very different immunological effects and some appear to be less adept in anti-inflammatory action than others, as ongoing research is now establishing.
The presence of symbiotic and pathogenic microorganisms in the gut or other peripheral tissues could lead to the maintenance of a pool of activated Tregs (both natural and inducible) that would maintain host immune homeostasis and enhance the threshold required for immune activation and induction of an immune response. The benefit of such deactivation is to decrease the instances of aberrant immune responses, such as allergic and autoimmune disorders. Pathogenic microorganisms may also have evolved to express antigens that cross-react with gut flora antigens. In infections, the removal or modification of the gut flora is associated with a modification of the phenotype of the host responses. Therefore, some microorganisms may hijack Tregs that are induced or activated in the gut to limit pathogenic responses against gut flora to ensure their own survival.
Over time, established GI infections may create a new homeostatic set point, in which reactivity to the chronic pathogen is minimized, with wider implications for responsiveness to self-antigens and allergens which may not be altogether detrimental. At this point, it remains unclear to what extent any recalibration of host immunity is induced purely by the pathogen, or by perturbation of the commensal population, or is a result of endogenous controls within the immune system itself. On the basis of both human and experimental studies discussed above, it seems likely that all three components play an essential role in reaching a stable and nonpathogenic steady state for the longer term.
Disclosure
None.
References
- 1.Smith DW, Nagler-Anderson C. Preventing intolerance: the induction of nonresponsiveness to dietary and microbial antigens in the intestinal mucosa. J Immunol. 2005;174:3851–7. doi: 10.4049/jimmunol.174.7.3851. [DOI] [PubMed] [Google Scholar]
- 2.Belkaid Y, Tarbell K. Regulatory T cells in the control of host–microorganism interactions. Annu Rev Immunol. 2009;27:551–89. doi: 10.1146/annurev.immunol.021908.132723. [DOI] [PubMed] [Google Scholar]
- 3.Izcue A, Coombes JL, Powrie F. Regulatory lymphocytes and intestinal inflammation. Annu Rev Immunol. 2009;27:313–38. doi: 10.1146/annurev.immunol.021908.132657. [DOI] [PubMed] [Google Scholar]
- 4.Macpherson AJ, Harris NL. Interactions between commensal intestinal bacteria and the immune system. Nat Rev Immunol. 2004;4:478–85. doi: 10.1038/nri1373. [DOI] [PubMed] [Google Scholar]
- 5.Vijay-Kumar A, Sanders CJ, Taylor RT, et al. Deletion of TLR5 results in spontaneous colitis in mice. J Clin Invest. 2007;117:3909–21. doi: 10.1172/JCI33084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.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]
- 7.Bashir ME, Louie S, Shi HN, Nagler-Anderson C. Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. J Immunol. 2004;172:6978–87. doi: 10.4049/jimmunol.172.11.6978. [DOI] [PubMed] [Google Scholar]
- 8.Sutmuller RP, den Brok MH, Kramer M, et al. Toll-like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest. 2006;116:485–94. doi: 10.1172/JCI25439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hall JA, Bouladoux N, Sun CM, 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]
- 10.Brenchley JM, Douek DC. HIV infection and the gastrointestinal immune system. Mucosal Immunol. 2008;1:23–30. doi: 10.1038/mi.2007.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lee J, Mo JH, Katakura K, 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]
- 12.Artis D. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nat Rev Immunol. 2008;8:411–20. doi: 10.1038/nri2316. [DOI] [PubMed] [Google Scholar]
- 13.Conte MP, Schippa S, Zamboni I, et al. Gut-associated bacterial microbiota in paediatric patients with inflammatory bowel disease. Gut. 2006;55:1760–7. doi: 10.1136/gut.2005.078824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kalliomaki M, Isolauri E. Role of intestinal flora in the development of allergy. Curr Opin Allergy Clin Immunol. 2003;3:15–20. doi: 10.1097/00130832-200302000-00003. [DOI] [PubMed] [Google Scholar]
- 15.Smits HH, Engering A, van der Kleij D, et al. Selective probiotic bacteria induce IL-10-producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin. J Allergy Clin Immunol. 2005;115:1260–7. doi: 10.1016/j.jaci.2005.03.036. [DOI] [PubMed] [Google Scholar]
- 16.Fujiwara D, Inoue S, Wakabayashi H, Fujii T. The anti-allergic effects of lactic acid bacteria are strain dependent and mediated by effects on both Th1/Th2 cytokine expression and balance. Int Arch Allergy Immunol. 2004;135:205–15. doi: 10.1159/000081305. [DOI] [PubMed] [Google Scholar]
- 17.Grangette C, Nutten S, Palumbo E, et al. Enhanced antiinflammatory capacity of a Lactobacillus plantarum mutant synthesizing modified teichoic acids. Proc Natl Acad Sci USA. 2005;102:10321–6. doi: 10.1073/pnas.0504084102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Gaboriau-Routhiau V, Rakotobe S, Lécuyer E, et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity. 2009;31:677–89. doi: 10.1016/j.immuni.2009.08.020. [DOI] [PubMed] [Google Scholar]
- 19.Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009:139. doi: 10.1016/j.cell.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Bach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med. 2002;347:911–20. doi: 10.1056/NEJMra020100. [DOI] [PubMed] [Google Scholar]
- 21.Wen L, Ley RE, Volchkov PY, 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]
- 22.O'Mahony C, Scully P, O'Mahony D, et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog. 2008;4:e1000112. doi: 10.1371/journal.ppat.1000112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Borchers AT, Selmi C, Meyers FJ, Keen CL, Gershwin ME. Probiotics and immunity. J gastroenterology. 2009;44:26–46. doi: 10.1007/s00535-008-2296-0. [DOI] [PubMed] [Google Scholar]
- 24.Di Giacinto C, Marinaro M, Sanchez M, Strober W, Boirivant M. Probiotics ameliorate recurrent Th1-mediated murine colitis by inducing IL-10 and IL-10-dependent TGF-β-bearing regulatory cells. J Immunol. 2005;174:3237–46. doi: 10.4049/jimmunol.174.6.3237. [DOI] [PubMed] [Google Scholar]
- 25.Zoumpopoulou G, Foligne B, Christodoulou K, Grangette C, Pot B, Tsakalidou E. Lactobacillus fermentum ACA-DC 179 displays probiotic potential in vitro and protects against trinitrobenzene sulfonic acid (TNBS)-induced colitis and Salmonella infection in murine models. Int J Food Microbiol. 2008;121:18–26. doi: 10.1016/j.ijfoodmicro.2007.10.013. [DOI] [PubMed] [Google Scholar]
- 26.Feleszko W, Jaworska J, Rha RD, et al. Probiotic-induced suppression of allergic sensitization and airway inflammation is associated with an increase of T regulatory-dependent mechanisms in a murine model of asthma. Clin Exp Allergy. 2007;37:498–505. doi: 10.1111/j.1365-2222.2006.02629.x. [DOI] [PubMed] [Google Scholar]
- 27.Karimi K, Inman MD, Bienenstock J, Forsythe P. Lactobacillus reuteri-induced regulatory T cells protect against an allergic airway response in mice. Am J Respir Crit Care Med. 2009;179:186–93. doi: 10.1164/rccm.200806-951OC. [DOI] [PubMed] [Google Scholar]
- 28.Foligne B, Zoumpopoulou G, Dewulf J, et al. A key role of dendritic cells in probiotic functionality. PLoS ONE. 2007;2:e313. doi: 10.1371/journal.pone.0000313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kullberg MC, Jankovic D, Gorelick PL, et al. Bacteria-triggered CD4+ T regulatory cells suppress Helicobacter hepaticus-induced colitis. J Exp Med. 2002;196:505–15. doi: 10.1084/jem.20020556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Heimesaat MM, Bereswill S, Fischer A, et al. Gram-negative bacteria aggravate murine small intestinal Th1-type immunopathology following oral infection with Toxoplasma gondii. J Immunol. 2006;177:8785–95. doi: 10.4049/jimmunol.177.12.8785. [DOI] [PubMed] [Google Scholar]
- 31.Noverr MC, Falkowski NR, McDonald RA, McKenzie AN, Huffnagle GB. Development of allergic airway disease in mice following antibiotic therapy and fungal microbiota increase: role of host genetics, antigen, and interleukin-13. Infect Immun. 2005;73:30–8. doi: 10.1128/IAI.73.1.30-38.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sullivan A, Edlund C, Nord CE. Effect of antimicrobial agents on the ecological balance of human microflora. Lancet Infect Dis. 2001;1:101–14. doi: 10.1016/S1473-3099(01)00066-4. [DOI] [PubMed] [Google Scholar]
- 33.Dethlefsen L, McFall-Ngai M, Relman DA. An ecological and evolutionary perspective on human–microbe mutualism and disease. Nature. 2007;449:811–18. doi: 10.1038/nature06245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Beaugerie L, Petit JC. Microbial–gut interactions in health and disease. Antibiotic-associated diarrhoea. Best Pract Res Clin Gastroenterol. 2004;18:337–52. doi: 10.1016/j.bpg.2003.10.002. [DOI] [PubMed] [Google Scholar]
- 35.Noverr MC, Huffnagle GB. The ‘microflora hypothesis’ of allergic diseases. Clin Exp Allergy. 2005;35:1511–20. doi: 10.1111/j.1365-2222.2005.02379.x. [DOI] [PubMed] [Google Scholar]
- 36.Prioult G, Nagler-Anderson C. Mucosal immunity and allergic responses: lack of regulation and/or lack of microbial stimulation? Immunol Rev. 2005;206:204–18. doi: 10.1111/j.0105-2896.2005.00277.x. [DOI] [PubMed] [Google Scholar]
- 37.Marra F, Lynd L, Coombes M, et al. Does antibiotic exposure during infancy lead to development of asthma? A systematic review and metaanalysis. Chest. 2006;129:610–8. doi: 10.1378/chest.129.3.610. [DOI] [PubMed] [Google Scholar]
- 38.Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6:e280. doi: 10.1371/journal.pbio.0060280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Pédron T, Sansonetti P. Commensals, bacterial pathogens and intestinal inflammation: an intriguing ménage à trois. Cell Host Microbe. 2008;3:344–7. doi: 10.1016/j.chom.2008.05.010. [DOI] [PubMed] [Google Scholar]
- 40.Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122:107–18. doi: 10.1016/j.cell.2005.05.007. [DOI] [PubMed] [Google Scholar]
- 41.Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453:620–5. doi: 10.1038/nature07008. [DOI] [PubMed] [Google Scholar]
- 42.Sun CM, Hall JA, Blank RB, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775–85. doi: 10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mucida D, Park Y, Kim G, 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]
- 44.Saurer L, McCullough KC, Summerfield A. In vitro induction of mucosa-type dendritic cells by all-trans retinoic acid. J Immunol. 2007;179:3504–14. doi: 10.4049/jimmunol.179.6.3504. [DOI] [PubMed] [Google Scholar]
- 45.Murai M, Turovskaya O, Kim G, et al. Interleukin 10 acts on regulatory T cells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis. Nat Immunol. 2009;10:1178–84. doi: 10.1038/ni.1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ardizzone S, Puttini PS, Cassinotti A, Porro GB. Extraintestinal manifestations of inflammatory bowel disease. Dig Liver Dis. 2008;40(Suppl. 2):S253–9. doi: 10.1016/S1590-8658(08)60534-4. [DOI] [PubMed] [Google Scholar]
- 47.Bleich A, Hopf S, Hedrich HJ, et al. Genetic dissection of granulomatous enterocolitis and arthritis in the intramural peptidoglycan–polysaccharide-treated rat model of IBD. Inflamm Bowel Dis. 2009;12:1794–802. doi: 10.1002/ibd.21018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fox JG, Beck P, Dangler CA, et al. Concurrent enteric helminth infection modulates inflammation and gastric immune responses and reduces Helicobacter-induced gastric atrophy. Nat Med. 2000;6:536–42. doi: 10.1038/75015. [DOI] [PubMed] [Google Scholar]
- 49.Elliott DE, Setiawan T, Metwali A, Blum A, Urban JF, Jr, Weinstock JV. Heligmosomoides polygyrus inhibits established colitis in IL-10-deficient mice. Eur J Immunol. 2004;34:2690–8. doi: 10.1002/eji.200324833. [DOI] [PubMed] [Google Scholar]
- 50.Bashir ME, Andersen P, Fuss IJ, Shi HN, Nagler-Anderson C. An enteric helminth infection protects against an allergic response to dietary antigen. J Immunol. 2002;169:3284–92. doi: 10.4049/jimmunol.169.6.3284. [DOI] [PubMed] [Google Scholar]
- 51.Metwali A, Setiawan T, Blum AM, et al. Induction of CD8+ regulatory T cells in the intestine by Heligmosomoides polygyrus infection. Am J Physiol Gastrointest Liver Physiol. 2006;291:G253–9. doi: 10.1152/ajpgi.00409.2005. [DOI] [PubMed] [Google Scholar]
- 52.Finney CAM, Taylor MD, Wilson MS, Maizels RM. Expansion and activation of CD4+CD25+ regulatory T cells in Heligmosomoides polygyrus infection. Eur J Immunol. 2007;37:1874–86. doi: 10.1002/eji.200636751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Rausch S, Huehn J, Kirchhoff D, et al. Functional analysis of effector and regulatory T cells in a parasitic nematode infection. Infect Immun. 2008;76:1908–19. doi: 10.1128/IAI.01233-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.D'Elia R, Behnke JM, Bradley JE, Else KJ. Regulatory T cells: a role in the control of helminth driven intestinal pathology and worm survival. J Immunol. 2009;182:2340–8. doi: 10.4049/jimmunol.0802767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Correale J, Farez M. Association between parasite infection and immune responses in multiple sclerosis. Ann Neurol. 2007;61:97–108. doi: 10.1002/ana.21067. [DOI] [PubMed] [Google Scholar]
- 56.Wilson MS, Taylor M, Balic A, Finney CAM, Lamb JR, Maizels RM. Suppression of allergic airway inflammation by helminth-induced regulatory T cells. J Exp Med. 2005;202:1199–212. doi: 10.1084/jem.20042572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Montagnoli C, Fallarino F, Gaziano R, et al. Immunity and tolerance to Aspergillus involve functionally distinct regulatory T cells and tryptophan catabolism. J Immunol. 2006;176:1712–23. doi: 10.4049/jimmunol.176.3.1712. [DOI] [PubMed] [Google Scholar]
- 58.Dunay IR, Damatta RA, Fux B, et al. Gr1(+) inflammatory monocytes are required for mucosal resistance to the pathogen Toxoplasma gondii. Immunity. 2008;29:306–17. doi: 10.1016/j.immuni.2008.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Curotto de Lafaille MA, Kutchukhidze N, Shen S, Ding Y, Yee H, Lafaille JJ. Adaptive Foxp3+ regulatory T cell-dependent and -independent control of allergic inflammation. Immunity. 2008;29:114–26. doi: 10.1016/j.immuni.2008.05.010. [DOI] [PubMed] [Google Scholar]
- 60.Schaub B, Lauener R, von Mutius E. The many faces of the hygiene hypothesis. J Allergy Clin Immunol. 2006;117:969–77. doi: 10.1016/j.jaci.2006.03.003. quiz 78. [DOI] [PubMed] [Google Scholar]
- 61.Su Z, Segura M, Morgan K, Loredo-Osti JC, Stevenson MM. Impairment of protective immunity to blood-stage malaria by concurrent nematode infection. Infect Immun. 2005;73:3531–9. doi: 10.1128/IAI.73.6.3531-3539.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Su Z, Segura M, Stevenson MM. Reduced protective efficacy of a blood-stage malaria vaccine by concurrent nematode infection. Infect Immun. 2006;74:2138–44. doi: 10.1128/IAI.74.4.2138-2144.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Urban JF, Jr, Steenhard NR, Solano-Aguilar GI, et al. Infection with parasitic nematodes confounds vaccination efficacy. Vet Parasitol. 2007;148:14–20. doi: 10.1016/j.vetpar.2007.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cooper PJ, Chico ME, Rodrigues LC, et al. Reduced risk of atopy among school-age children infected with geohelminth parasites in a rural area of the tropics. J Allergy Clin Immunol. 2003;111:995–1000. doi: 10.1067/mai.2003.1348. [DOI] [PubMed] [Google Scholar]
- 65.Dagoye D, Bekele Z, Woldemichael K, et al. Wheezing, allergy, and parasite infection in children in urban and rural Ethiopia. Am J Respir Crit Care Med. 2003;167:1369–73. doi: 10.1164/rccm.200210-1204OC. [DOI] [PubMed] [Google Scholar]
- 66.Summers RW, Elliott DE, Urban JF, Jr, Thompson RA, Weinstock JV. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology. 2005;128:825–32. doi: 10.1053/j.gastro.2005.01.005. [DOI] [PubMed] [Google Scholar]
- 67.Fallon PG, Mangan NE. Suppression of Th2-type allergic reactions by helminth infection. Nat Rev Immunol. 2007;7:220–30. doi: 10.1038/nri2039. [DOI] [PubMed] [Google Scholar]