Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Aug 14.
Published in final edited form as: Curr Opin Microbiol. 2016 Apr 23;32:14–18. doi: 10.1016/j.mib.2016.04.004

Macrobiota — helminths as active participants and partners of the microbiota in host intestinal homeostasis

William C Gause 1, Rick M Maizels 2
PMCID: PMC4983462  NIHMSID: NIHMS801660  PMID: 27116368

Abstract

Important insights have recently been gained in our understanding of the intricate relationship in the intestinal milieu between the vertebrate host mucosal immune response, commensal bacteria, and helminths. Helminths are metazoan worms (macrobiota) and trigger immune responses that include potent regulatory components capable of controlling harmful inflammation, protecting barrier function and mitigating tissue damage. They can secrete a variety of products that directly affect immune regulatory function but they also have the capacity to influence the composition of microbiota, which can also then impact immune function. Conversely, changes in microbiota can affect susceptibility to helminth infection, indicating that crosstalk between these two disparate groups of endobiota can play an essential role in host intestinal immune function and homeostasis.


Intestinal commensal bacteria and helminths flourish in vertebrate hosts, an outcome of a coevolutionary dynamic that has persisted for several hundred million years. Increasing evidence suggests that this three-way partnership has resulted in complex adaptations that have shaped the physiology of each of these very different organisms in health and disease. As a result, homeostasis in vertebrates may now require the presence of both commensal microbiota and macrobiota, including helminths. The absence of either of these organisms may dispose towards a dysregulated immune system, which may favor harmful inflammatory responses that can contribute to a variety of disease states.

For both commensal bacteria and helminths, essential adaptations promote acceptance by the vertebrate host. Shared mechanisms may include immune evasion strategies, such as molecular mimicry, activation of immune regulatory pathways, or diversionary stimulation of ineffective immune responses. The evolutionary response of the vertebrate host has been to develop suites of resistance mechanisms to control and eradicate the invading organism. A fascinating strategy is where the host opts to mitigate adverse effects of infection, accommodating an organism but minimizing pathology (a non-immunological form of tolerance [1]). Such tolerance adaptations decrease the impact of the invading organism without actually reducing the burden.

Tolerance mechanisms may include both control of harmful inflammation and enhanced wound healing that together mitigate organ and tissue damage. Tolerance mechanisms may enhance fitness for the invasive organism as well as for the vertebrate host. As such, enhanced tolerance may result from combined contributions of the vertebrate host, microbiota, and macrobiota. Recent studies also suggest that commensal bacteria and helminths interact providing signals that impact their survival in the vertebrate host. Thus, there appears to be a three way multilateral partnership that supports coexistence of these quite different organisms. In this review, we will discuss recent studies elucidating how their interactions may impact health and disease.

A number of studies published over the last few years now indicate that helminth infection can alter the composition of the intestinal microbiome with respect to both species abundance and composition [2]. For example, 3 independent laboratories reported that infection with the mouse duodenal parasite Heligmosomoides polygyrus expanded the proportion of Lactobacillaceae and Enterobacteriaceae in the gut [35]. Moreover, while chronic infection with the mouse whipworm Trichuris muris similarly raised Lacto-bacillaceae representation, it also reduced overall microbiota diversity, a factor often associated with poorer homeostatic control, reflecting an ecological imbalance in the intestinal microbial community [6,7]. Intriguingly, removal of parasites restored the ‘naïve’ flora observed in uninfected mice, suggesting that helminth-induced changes in the microbiota are reversible by clearance of the macrobiota [7]. Likewise, in wild mice (Apodemus flavicollis) a correlation was observed between increased bacterial microbiota richness and helminth infections and different types of helminths elicited characteristic changes in the composition and abundance of microbiota species [8]. In humans, reports are only now emerging and involve very different sets of helminth-exposed communities (summarised in [2]); while in a Malaysian population, microbial diversity was greater in those infected with parasites [9], it is not yet clear if this is a pattern that will be found generally applicable [6].

Taken together, these studies demonstrate that parasite infection can change the composition of gut microbiota, but do not address the possibility that changes in the microbiome may also affect susceptibility of the host to parasite infection. However, two laboratories have indeed shown that introducing higher levels of Lactobacillaceae microbes can increase susceptibility to helminth infection [5,10], raising the suggestion that helminths and certain commensal species may mutually reinforce each other’s presence. The reduced susceptibility of germ-free mice to helminth infection [11] lends further support to this proposition. Recent studies further suggest that intestinal microbiota may also affect immune responses to helminths. In a murine model of schistosomiasis, antibiotic-mediated depletion of gut bacteria significantly reduced intestinal inflammation and decreased intestinal granuloma development [8,12,13].

What may be the mechanisms underlying these effects? In the case of intestinal helminths, an important factor is the physical disruption to the epithelial surface often involving a barrier breach and causing bacterial translocation. Th1/Th17 responses evoked by opportunistic bacterial exposure may dampen the Th2 mode of immunity required for parasite expulsion, as suggested by the heightened resistance of MyD88-deficient mice to intestinal helminths [14,15]. Conversely, helminths may dampen inflammatory responses to bacteria as shown in a remarkable study of idiopathic bowel disease in captive macaques who, when given Trichuris worm parasites, showed a shift to a counter-inflammatory Th2-dominated environment in which microbial dysregulation is reversed and barrier function is restored [16].

Both helminths and the microbiota are frequently linked to expanded regulatory T cell (Treg) activity [17,18], and mice in which the ability of commensals to induce intestinal Tregs is compromised were found to be more resistant to H. polygyrus infection [19]. More broadly, the stimulation of Treg activity has emerged as a central explanation for the beneficial effects of certain probiotic bacteria, and controlled helminth infection, in ameliorating inflammatory diseases such as allergy and autoimmune disorders [20]. An important question now arises, of whether these changes in regulation of the immune response are caused by direct effects of the helminth parasite (e.g. by, production of excretory/secretory products) or are instead an indirect effect of the altered microbiome.

Recent studies raise the possibility that the latter may in fact be an important contributor to helminth-induced immune regulation. In one report, mice were infected with H. polygyrus, which is known to activate regulatory T cells capable of mediating protection against allergic asthma [21] resulting in this case in increased abundance of bacteria belonging to Clostridiales. Remarkably, transfer of microbiota-rich intestinal contents from infected mice was sufficient to trigger immune regulatory populations capable of ameliorating allergic asthma in uninfected recipient mice. Further analyses showed that short chain fatty acids (SCFAs), produced by these intestinal bacteria, was essential for increased Treg cell suppressor activation and the associated production of anti-inflammatory cytokines that controlled asthma in H. polygyrus-infected mice [22]. Interestingly, helminths can also produce SCFAs [23], while the intestinal lumen can carry many host products such as cytokines, exosomes and even micro-RNAs [24], raising other possibilities for how each partner in the host–parasite–commensal triangle may influence the outcome of the immune response.

The extent to which helminths may, like the microbiota [25], influence the metabolic status of their host has only recently been questioned. T. muris-infected mice were shown to exhibit extensive changes in fecal metabolomic products [7], although in this and other studies it remains to be determined which changes are the result of microbial compositional and biosynthetic alterations consequent upon the nematode infection.

Helminths can in any case directly modulate the vertebrate host immune response through a number of intricate mechanisms, many of which are likely to indirectly impact the microbial cohabitants. As these large multicellular parasites migrate through tissues, they cause cellular damage and release of danger signals, such as trefoil factor 2 (tff2) and adenosine, which can in turn trigger production of IL-33, IL-25 and TSLP, inducing the release of key type 2 cytokines, including IL-4 and IL-13 [26,27]. Through a positive feedback circuit, IL-4/ 13 induces expansion of epithelial tuft cells, the source of IL-25, which drives further IL-13 production from both innate and adaptive lymphocytes that can mediate worm expulsion [28]. Increased IL-13 can also enhance mucous production, and a switch from Muc2 to Muc5ac that is necessary for resistance to infection [29], most probably also changing the microbial environment in the intestine.

Type 2 immunity also includes differentiation of alternatively activated (M2) macrophages and their production of factors important in tissue repair, such as RELMα [30] and insulin-like growth factor (IGF-1) [31]. Amphiregulin is also upregulated and produced by a variety of cells including epithelial cells, innate lymphoid cells and T reg cells, and may also enhance T reg cell function [32]. As discussed above, production of type 2 cytokines and activation of Treg cells may in turn dampen type 1 inflammatory responses to intestinal bacteria by modulating both TLR signaling and the production of type 1 cytokines, including IFN-γ. Polarisation of the response in helminth infection also raises levels of the antimicrobial products angiogenin 4 [33] and RegIIIγ in the intestines of mice [34].

Taken together, the type 2 response and the M2 macrophages it induces can mitigate tissue damage associated with helminth infection and as such may substantially enhance tolerance of these eukaryotic pathogens by the vertebrate host [35]. M2 macrophages, induced through IL-4R signaling, can also mediate resistance to helminths through products such as Arginase-1 [36,37]. In this respect, the type 2 response that has evolved in the mammalian immune system can be seen to have resolved the conflict between tolerance and resistance as the same pathway mediates parasite killing as well as necessary tissue repair [38].

A further, and unexpected, feature of the type 2 response is the degree of innate memory which is established through cell phenotypes, which persist for long periods in the host thereby contributing to the memory response upon subsequent exposure [39]. This can help explain how helminth infections may rebalance immune homeostasis by shifting the setpoint away from harmful inflammatory responses associated with type 1 immunity, and provide a new long-term context for the recent description of ‘trained immunity’ [40].

Helminths also have the capacity to release excretory/ secretory (ES) products that can modulate immune function, a likely consequence of host:parasite coevolution. Indeed, the ability of helminth ES to recapitulate much of the suppressive impact of live parasite infection [41] is the strongest evidence that helminth immune modulation is largely a result of direct interactions with the host, although indirect effects via microbial changes will surely play a part. While ES products are primarily thought to enhance helminth fitness by downregulating protective immune responses, it is likely that they also promote tolerance mechanisms to minimize mortality of their host and insulate their own niche from inflammatory reactions. Thus, the ES of H. polygyrus includes a functional mimic of the most tolerogenic mammalian cytokine, TGFβ, able to induce Treg differentiation [42], as well as a separate activity which inhibits pro-inflammatory responses of dendritic cells to TLR ligand exposure [43]. Most recently, the discovery that intestinal helminth parasites release extracellular vesicles, or exosomes, loaded with both proteins and micro-RNAs, which down-regulate (in the case of H. polygyrus) the IL-33R [44] opens up new pathways of communication between macrobiota, commensals and the mammalian host [45].

Finally, a relatively little-explored question is whether helminths may act directly on their intestinal microbial neighbors to regulate their populations; for example, they may disrupt the bacterial niche, preferentially deplete essential nutrients, or even release anti-bacterial products. Recent work has discovered a suite of antimicrobial mechanisms which defend free-living nematodes such as Caenorhabditis elegans from bacterial invasion [46], so it is plausible that species evolving in the mammalian intestine have adapted these mechanisms to control the microbiome. For example, among the ES proteins secreted by the luminal-dwelling adult stage of H. polygyrus are at least 8 lysozymes with potential antibacterial activity [47], as well as many small polypeptides that could include defensin-like products. Future work may well, therefore, identify novel mediators for manipulating the microbiome that could promote the anti-inflammatory effects of both helminth and beneficial commensal species.

In conclusion, it should be noted that humans are thought to carry up to 1000 different bacterial species, as well as an intestinal virome that is only now being characterized [48]. In contrast, the vast majority of intestinal helminth infections of humans are accounted for by a handful of species such as Ascaris, hookworms, Strongyloides, Taenia and Trichuris. Evidently, these successful parasites have each evolved unique strategies to manipulate both the host and its microbial constituents to remarkable effect, through pathways that are only now coming to light.

Acknowledgments

WCG is supported by grants from the Amelior Foundation and the National Institutes of Health (1R01AI107588). RMM is supported by grants from the Rainin Foundation (Ref. 12-H4) and the Wellcome Trust (Ref. 106122).

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

• of special interest

•• of outstanding interest

  • 1.Medzhitov R, Schneider DS, Soares MP. Disease tolerance as a defense strategy. Science. 2012;335:936–941. doi: 10.1126/science.1214935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Reynolds LA, Finlay BB, Maizels RM. Cohabitation in the intestine: interactions among helminth parasites, bacterial microbiota, and host immunity. J Immunol. 2015;195:4059–4066. doi: 10.4049/jimmunol.1501432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Walk ST, Blum AM, Ewing SA, Weinstock JV, Young VB. Alteration of the murine gut microbiota during infection with the parasitic helminth Heligmosomoides polygyrus. Inflamm Bowel Dis. 2010;16:1841–1849. doi: 10.1002/ibd.21299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Rausch S, Held J, Fischer A, et al. Small intestinal nematode infection of mice is associated with increased enterobacterial loads alongside the intestinal tract. PLOS ONE. 2013;8:e74026. doi: 10.1371/journal.pone.0074026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5•.Reynolds LA, Smith KA, Filbey KJ, et al. Commensal–pathogen interactions in the intestinal tract: lactobacilli promote infection with, and are promoted by, helminth parasites. Gut Microbes. 2014;5:522–532. doi: 10.4161/gmic.32155. This paper, alone with Ref. 10, show that certain Lactobacillus species render the host more susceptible to helminth infection, and in this paper, that the helminth H. polygyrus promotes outgrowth of the Lactobacillus that favors it. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Holm JB, Sorobetea D, Kiilerich P, et al. Chronic Trichuris muris infection decreases diversity of the intestinal microbiota and concomitantly increases the abundance of lactobacilli. PLOS ONE. 2015;10:e0125495. doi: 10.1371/journal.pone.0125495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7•.Houlden A, Hayes KS, Bancroft AJ, et al. Chronic Trichuris muris infection in C57BL/6 mice causes significant changes in host microbiota and metabolome: effects reversed by pathogen clearance. PLOS ONE. 2015;10:e0125945. doi: 10.1371/journal.pone.0125945. This paper catalogues the dynamic changes to the microbiota following an intestinal helminth infection, coupled with alterations in metabolic breakdown of nutrients; significantly, following drug-mediated clearance, the microbiome slowly reverts to the steady-state, confirming also that helminths are causative agents in determining microbial composition. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kreisinger J, Bastien G, Hauffe HC, Marchesi J, Perkins SE. Interactions between multiple helminths and the gut microbiota in wild rodents. Philos Trans R Soc Lond Ser B Biol Sci. 2015:370. doi: 10.1098/rstb.2014.0295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lee SC, Tang MS, Lim YA, et al. Helminth colonization is associated with increased diversity of the gut microbiota. PLoS Negl Trop Dis. 2014;8:e2880. doi: 10.1371/journal.pntd.0002880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dea-Ayuela MA, Rama-Iñiguez S, Bolás-Fernandez F. Enhanced susceptibility to Trichuris muris infection of B10Br mice treated with the probiotic Lactobacillus casei. Int Immunopharmacol. 2008;8:28–35. doi: 10.1016/j.intimp.2007.10.003. [DOI] [PubMed] [Google Scholar]
  • 11.Chang J, Wescott RB. Infectivity, fecundity, and survival of Nematospiroides dubius in gnotobiotic mice. Exp Parasitol. 1972;32:327–334. doi: 10.1016/0014-4894(72)90060-4. [DOI] [PubMed] [Google Scholar]
  • 12.Holzscheiter M, Layland LE, Loffredo-Verde E, et al. Lack of host gut microbiota alters immune responses and intestinal granuloma formation during schistosomiasis. Clin Exp Immunol. 2014;175:246–257. doi: 10.1111/cei.12230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.McKenney EA, Williamson L, Yoder AD, Rawls JF, Bilbo SD, Parker W. Alteration of the rat cecal microbiome during colonization with the helminth Hymenolepis diminuta. Gut Microbes. 2015;6:182–193. doi: 10.1080/19490976.2015.1047128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Helmby H, Grencis RK. Essential role for TLR4 and MyD88 in the development of chronic intestinal nematode infection. Eur J Immunol. 2003;33:2974–2979. doi: 10.1002/eji.200324264. [DOI] [PubMed] [Google Scholar]
  • 15.Reynolds LA, Harcus Y, Smith KA, et al. MyD88 signaling inhibits protective immunity to the gastrointestinal helminth parasite Heligmosomoides polygyrus. J Immunol. 2014;193:2984–2993. doi: 10.4049/jimmunol.1401056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16•.Broadhurst MJ, Ardeshir A, Kanwar B, et al. Therapeutic helminth infection of macaques with idiopathic chronic diarrhea alters the inflammatory signature and mucosal microbiota of the colon. PLoS Pathog. 2012;8:e1003000. doi: 10.1371/journal.ppat.1003000. This paper presents a comprehensive analysis in primates of the impact of helminth infection, linking the anti-inflammatory effects of Trichuris with suppression of the Th1 response and alterations in both microbial diversity and the ability of microbes to attach to the intestinal epithelium. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Geuking MB, Cahenzli J, Lawson MA, et al. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity. 2011;34:794–806. doi: 10.1016/j.immuni.2011.03.021. [DOI] [PubMed] [Google Scholar]
  • 18.Maizels RM, Smith KA. Regulatory T cells in infection. Adv Immunol. 2011;112:73–136. doi: 10.1016/B978-0-12-387827-4.00003-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19•.Ohnmacht C, Park JH, Cording S, et al. The microbiota regulates type 2 immunity through RORgammat+ T cells. Science. 2015;349:989–993. doi: 10.1126/science.aac4263. This paper proposes that intestinal microbe induction of RORγt in T cells is accompanied, in the presence of retinoic acid, by generation of RORγt+ Tregs. In the absence of this specific Treg subset, Th2 responses become exuberent, aggravating Type 2 colitis, but also rendering mice more resistant to the intestinal helminth H. polygyrus. [DOI] [PubMed] [Google Scholar]
  • 20.McSorley HJ, Maizels RM. Helminth infections and host immune regulation. Clin Microbiol Rev. 2012;25:585–608. doi: 10.1128/CMR.05040-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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;202:1199–1212. doi: 10.1084/jem.20042572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zaiss MM, Rapin A, Lebon L, et al. The intestinal microbiota contributes to the ability of helminths to modulate allergic inflammation. Immunity. 2015;43:998–1010. doi: 10.1016/j.immuni.2015.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tielens AGM, van Grinsven KWA, Henze K, van Hellemond JJ, Martin W. Acetate formation in the energy metabolism of parasitic helminths and protists. Int J Parasitol. 2010;40:387–397. doi: 10.1016/j.ijpara.2009.12.006. [DOI] [PubMed] [Google Scholar]
  • 24.Liu S, da Cunha AP, Rezende RM, et al. The host shapes the gut microbiota via fecal MicroRNA. Cell Host Microbe. 2016;19:32–43. doi: 10.1016/j.chom.2015.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Turnbaugh PJ, Backhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3:213–223. doi: 10.1016/j.chom.2008.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wills-Karp M, Rani R, Dienger K, et al. Trefoil factor 2 rapidly induces interleukin 33 to promote type 2 immunity during allergic asthma and hookworm infection. J Exp Med. 2012;209:607–622. doi: 10.1084/jem.20110079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27•.Patel N, Wu W, Mishra PK, et al. A2B adenosine receptor induces protective antihelminth type 2 immune responses. Cell Host Microbe. 2014;15:339–350. doi: 10.1016/j.chom.2014.02.001. This paper, along with reference 26, were among the first to identify danger associated molecular patterns (DAMPS) essential in triggering type 2 immune responses to helminths. [DOI] [PubMed] [Google Scholar]
  • 28.Gerbe F, Sidot E, Smyth DJ, et al. Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites. Nature. 2016;529:226–230. doi: 10.1038/nature16527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hasnain SZ, Evans CM, Roy M, et al. Muc5ac: a critical component mediating the rejection of enteric nematodes. J Exp Med. 2011;208:893–900. doi: 10.1084/jem.20102057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Knipper JA, Willenborg S, Brinckmann J, et al. Interleukin-4 receptor alpha signaling in myeloid cells controls collagen fibril assembly in skin repair. Immunity. 2015;43:803–816. doi: 10.1016/j.immuni.2015.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chen F, Liu Z, Wu W, et al. An essential role for TH2-type responses in limiting acute tissue damage during experimental helminth infection. Nat Med. 2012;18:260–266. doi: 10.1038/nm.2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zaiss DM, Yang L, Shah PR, Kobie JJ, Urban JF, Mosmann TR. Amphiregulin, a TH2 cytokine enhancing resistance to nematodes. Science. 2006;314:1746. doi: 10.1126/science.1133715. [DOI] [PubMed] [Google Scholar]
  • 33.D’Elia R, DeSchoolmeester ML, Zeef LA, Wright SH, Pemberton AD, Else KJ. Expulsion of Trichuris muris is associated with increased expression of angiogenin 4 in the gut and increased acidity of mucins within the goblet cell. BMC Genomics. 2009;10:492. doi: 10.1186/1471-2164-10-492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Su L, Su CW, Qi Y, et al. Coinfection with an intestinal helminth impairs host innate immunity against Salmonella enterica serovar Typhimurium and exacerbates intestinal inflammation in mice. Infect Immun. 2014;82:3855–3866. doi: 10.1128/IAI.02023-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gause WC, Wynn TA, Allen JE. Type 2 immunity and wound healing: evolutionary refinement of adaptive immunity by helminths. Nat Rev Immunol. 2013;13:607–614. doi: 10.1038/nri3476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Anthony RM, Urban JF, Jr, Alem F, et al. Memory TH2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat Med. 2006;12:955–960. doi: 10.1038/nm1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Obata-Ninomiya K, Ishiwata K, Tsutsui H, et al. The skin is an important bulwark of acquired immunity against intestinal helminths. J Exp Med. 2013 doi: 10.1084/jem.20130761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Allen JE, Maizels RM. Diversity and dialogue in immunity to helminths. Nat Rev Immunol. 2011;11:375–388. doi: 10.1038/nri2992. [DOI] [PubMed] [Google Scholar]
  • 39•.Chen F, Wu W, Millman A, et al. Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion. Nat Immunol. 2014;15:938–946. doi: 10.1038/ni.2984. This paper showed that macrophages can mediated acquired resistance to helminths suggesting that trained innate immunity can contribute to the type 2 memory immune response. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Netea MG, Latz E, Mills KH, O’Neill LA. Innate immune memory: a paradigm shift in understanding host defense. Nat Immunol. 2015;16:675–679. doi: 10.1038/ni.3178. [DOI] [PubMed] [Google Scholar]
  • 41.Hewitson JP, Grainger JR, Maizels RM. Helminth immunoregulation: the role of parasite secreted proteins in modulating host immunity. Mol Biochem Parasitol. 2009;167:1–11. doi: 10.1016/j.molbiopara.2009.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Grainger JR, Smith KA, Hewitson JP, et al. Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-β pathway. J Exp Med. 2010;207:2331–2341. doi: 10.1084/jem.20101074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Segura M, Su Z, Piccirillo C, Stevenson MM. Impairment of dendritic cell function by excretory–secretory products: a potential mechanism for nematode-induced immunosuppression. Eur J Immunol. 2007;37:1887–1904. doi: 10.1002/eji.200636553. [DOI] [PubMed] [Google Scholar]
  • 44.Buck AH, Coakley G, Simbari F, et al. Exosomes secreted by nematode parasites transfer small RNAs to mammalian cells and modulate innate immunity. Nat Commun. 2014;5:5488. doi: 10.1038/ncomms6488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Coakley G, Maizels RM, Buck AH. Exosomes and other extracellular vesicles: the new communicators in parasite infections. Trends Parasitol. 2015;31:477–489. doi: 10.1016/j.pt.2015.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ewbank JJ, Zugasti O. C elegans: model host and tool for antimicrobial drug discovery. Dis Models Mech. 2011;4:300–304. doi: 10.1242/dmm.006684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hewitson JP, Harcus Y, Murray J, et al. Proteomic analysis of secretory products from the model gastrointestinal nematode Heligmosomoides polygyrus reveals dominance of Venom Allergen-Like (VAL) proteins. J Proteomics. 2011;74:1573–1594. doi: 10.1016/j.jprot.2011.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Norman JM, Handley SA, Baldridge MT, et al. Disease-specific alterations in the enteric virome in inflammatory bowel disease. Cell. 2015;160:447–460. doi: 10.1016/j.cell.2015.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES