Elsevier

Experimental Parasitology

Volume 158, November 2015, Pages 8-17
Experimental Parasitology

Full length article
Suppression of OVA-alum induced allergy by Heligmosomoides polygyrus products is MyD88-, TRIF-, regulatory T- and B cell-independent, but is associated with reduced innate lymphoid cell activation

https://doi.org/10.1016/j.exppara.2015.02.009Get rights and content

Highlights

  • H. polygyrus ES (HES) suppresses alum-induced type 2 responses in MyD88xTRIF doubly-deficient mice.

  • HES does not require the presence of B cells to block alum-induced type 2 responses.

  • Regulatory T cell depletion does not reverse suppression of alum-induced type 2 responses by HES.

  • HES-administered mice show significant inhibition of type 2 innate lymphoid cell activation.

Abstract

The murine intestinal nematode Heligmosomoides polygyrus exerts multiple immunomodulatory effects in the host, including the suppression of allergic inflammation in mice sensitized to allergen presented with alum adjuvant. Similar suppression is attained by co-administration of H. polygyrus excretory/secretory products (HES) with the sensitizing dose of ovalbumin (OVA) in alum. We investigated the mechanism of suppression by HES in this model, and found it was maintained in MyD88xTRIF-deficient mice, implying no role for helminth- or host-derived TLR ligands, or IL-1 family cytokines that signal in a MyD88- or TRIF-dependent manner. We also found suppression was unchanged in µMT mice, which lack B2 B cells, and that suppression was not abrogated when regulatory T cells were depleted in Foxp3.LuciDTR-4 mice. However, reduced IL-5 production was seen in the first 12 h after injection of OVA-alum when HES was co-administered, associated with reduced activation of IL-5+ and IL-13+ group 2 innate lymphoid cells. Thus, the suppressive effects of HES on alum-mediated OVA sensitization are reflected in the very earliest innate response to allergen exposure in vivo.

Introduction

Both epidemiological and experimental studies have established a consistent negative correlation between parasitic helminth infections and the incidence of allergic reactivity (Araujo, de Carvalho, 2006, Cooper, 2009, McSorley, Maizels, 2012, Yazdanbakhsh et al, 2002). These observations have led both to a revised “hygiene hypothesis” – the proposition that in the absence of infections, the immune system is prone to hyperactivity (Maizels et al, 2014, Yazdanbakhsh et al, 2002) – and to “helminth therapy”, the concept of using parasitic organisms as therapeutic agents against immune-mediated diseases (Evans, Mitre, 2014, Feary et al, 2010, Weinstock, Elliott, 2013). Furthermore, vaccination responses, dependent on effective Th2-mediated antibody production, have been shown to be less effective in parasite-endemic areas (Cooper et al, 1999, Stelekati, Wherry, 2012). Hence it is widely thought that parasites release immunomodulatory products tailored to manipulate host immune responses, especially the type 2/Th2 pathway which generates both anti-helminth immunity and allergy (Danilowicz-Luebert et al, 2011, Finlay et al, 2014, Johnston et al, 2009, Maizels, Yazdanbakhsh, 2003). In order to more completely define the molecular motifs and immune mechanisms by which helminths modulate type 2 responses, we and others have used the products of helminth parasites, administered into uninfected mice, to replicate the beneficial effects of infection (Ebner et al, 2014, McSorley et al, 2012, McSorley et al, 2014, Rzepecka et al, 2013, Schabussova et al, 2013, Trujillo-Vargas et al, 2007, Yang et al, 2007).

The mouse intestinal nematode parasite, Heligmosomoides polygyrus, exerts a broad range of immunomodulatory effects in the infected host, including suppression of allergy, autoimmunity and colitis (Maizels et al., 2012). Many of these effects can be recapitulated by soluble H. polygyrus excretory/secretory products (HES), while in vitro HES is able to suppress dendritic cell maturation (Segura et al., 2007) and to induce regulatory T cells (Tregs) through the TGF-βR pathway (Grainger et al., 2010). In the context of allergy, we recently showed that HES could replicate the suppressive effects of H. polygyrus infection, when administered at sensitization in asthma models driven by Alternaria fungal extract (McSorley et al., 2014) or ovalbumin (OVA) co-precipitated onto alum adjuvant (McSorley et al., 2012). In the Alternaria model of asthma, fungal allergens are introduced directly to the airways without recourse to exogenous adjuvant (Kobayashi et al., 2009). In this setting, HES pre-empts adaptive immune responses by suppressing the release of IL-33 immediately after administration of allergen into the lungs. This led to reduced group 2 innate lymphoid cell (ILC2) activation and reduced subsequent T cell-mediated inflammation in the lungs (McSorley et al., 2014). In the OVA-alum model of asthma, mice are sensitized by intraperitoneal immunization of OVA with the type-2 promoting adjuvant alum (Lloyd, 2007, Oleszycka, Lavelle, 2014). In this model, a mechanism of suppression by HES has not yet been identified.

TLR ligands, such as bacterial endotoxin, when co-administered at sensitization with OVA-alum, can result in suppression of subsequent immune responses at challenge (Bortolatto et al, 2008, Eisenbarth, 2008). Although HES from our laboratory contained below-threshold levels of endotoxin contamination, helminth secretions are known to contain other TLR ligands (van der Kleij et al., 2002), which potentially could act in a similar manner to suppress responses at sensitization. Furthermore, HES has recently been shown to induce the release of IL-1β from macrophages, resulting in the suppression of pro-allergenic IL-25 and IL-33 responses, and a diminished Th2 response (Zaiss et al., 2013a). Of note, for functional TLR and IL-1R responses, both of the signaling molecules MyD88 and TRIF are required, although alum-induced allergic responses are intact in mice lacking either or both of these adapter proteins (Eisenbarth et al, 2008, Gavin et al, 2006, Piggott et al, 2005).

In H. polygyrus infection, Tregs expand in the draining mesenteric lymph nodes, and can, when transferred to naive mice, suppress allergic airway disease (Wilson et al, 2005, Zaiss et al, 2013b). Likewise, Tregs induced in vitro by HES or mammalian TGF-β can be transferred into uninfected mice, and also suppress allergic immune responses in their adoptive hosts (Grainger et al., 2010). When we investigated the effects of HES in mice receiving OVA-alum injections, however, we found reduced rather than greater Treg numbers in the lung; moreover, administration of recombinant mammalian TGF-β with equivalent activity to the TGF-β mimic in HES could not replicate suppression (McSorley et al., 2012). These data argued against, but did not exclude, a role for Treg induction in suppression of asthmatic responses by HES in the OVA-alum model.

Previous work by Wilson et al. showed that infection with H. polygyrus also results in a regulatory B cell population able to suppress allergic airway disease (Wilson et al., 2010). These B cells expressed follicular B2 B cell markers, with suppression independent of IL-10. Regulatory B cells induced by schistosomes have also been shown to suppress responses in models of asthma (Amu et al, 2010, Smits et al, 2007, van der Vlugt et al, 2012) suggesting that the ability of parasites to drive regulatory B cells may be a common mechanism of asthma suppression. The effect on B cells is further emphasized by the potent suppression of B cell antibody responses observed in mice given HES with OVA-alum (McSorley et al., 2012), although the role of regulatory B cells had not yet been investigated in this model.

Here, we test the hypotheses that HES could be suppressing alum-driven allergic immune responses through TLR or IL-1-family members signaling through MyD88 or TRIF, or regulatory B cell (Breg) or Treg induction. Using MyD88xTRIF double-deficient mice, we found that suppression was not associated with TLR or IL-1 family members signaling through these adaptor proteins. Furthermore, using µMT and Foxp3.LuciDTR-4 mice, we found that suppression was unaffected in the absence of B2 B cells or with depletion of regulatory T cells. Instead we found that HES co-administration was associated with reduced early (<12 h post-administration) production of type 2 cytokines by group 2 innate lymphoid cells at the site of administration.

Section snippets

Parasites and reagents

The life cycle of H. polygyrus bakeri was maintained, and HES products were prepared, as described elsewhere (Johnston et al., 2015). Class IV Ovalbumin was purchased from Sigma, Gillingham, Dorset, UK.

Mice

BALB/c, C57BL/6, MyD88xTRIF-double deficient (Uematsu et al., 2002), µMT (Kitamura et al., 1991) and Foxp3.LuciDTR-4 (Suffner et al., 2010) mice were bred in-house at the University of Edinburgh and accommodated according to Home Office regulations.

OVA-alum model

Induction of airway allergic inflammation was

OVA-alum induced airway allergy, and its suppression by HES, do not require MyD88 or TRIF signaling

MyD88, together with TRIF, are central adapter molecules which transmit signals from cell receptors for Toll-like ligands and IL-1 family cytokines (Boraschi and Tagliabue, 2013). To evaluate if these pathways are involved in HES-driven suppression of the allergic response to OVA-alum sensitization, we made use of MyD88xTRIF double-deficient mice, which (except in rare cases (Davis et al, 2006, Santos-Sierra et al, 2009)) cannot respond to ligation of TLR and IL-1 family receptors (Yamamoto

Discussion

The products of helminth parasites have extraordinary capability to suppress a broad spectrum of immune responses to model immunogens, allergens, autoantigens and vaccines (Adisakwattana et al, 2009, Harnett, 2014, Johnston et al, 2009, Maizels et al, 2014, McSorley, Maizels, 2012). We previously showed that H. polygyrus HES strongly inhibits pathology in the OVA-alum model of asthma, in which OVA-specific Th2 responses are induced by systemic injection of OVA protein adsorbed to an alum

Acknowledgements

We gratefully acknowledge funding from the American Asthma Foundation (all authors, grant number 10-0234), Asthma UK (HMcS, grant number AUK-SPD-2012-172) and the Wellcome Trust (RMM, grant number 090281).

References (75)

  • C.J. Oliphant et al.

    MHCII-mediated dialog between group 2 innate lymphoid cells and CD4+ T cells potentiates type 2 immunity and promotes parasitic helminth expulsion

    Immunity

    (2014)
  • J. Rzepecka et al.

    The helminth product, ES-62, protects against airway inflammation by resetting the Th cell phenotype

    Int. J. Parasitol

    (2013)
  • H.H. Smits et al.

    Protective effect of Schistosoma mansoni infection on allergic asthma depends on intensity and chronicity of infection

    J. Allergy Clin. Immunol

    (2007)
  • E. Stelekati et al.

    Chronic bystander infections and immunity to unrelated antigens

    Cell Host Microbe

    (2012)
  • D. Togbe et al.

    Nonredundant roles of TIRAP and MyD88 in airway response to endotoxin, independent of TRIF, IL-1 and IL-18 pathways

    Lab. Invest

    (2006)
  • D. van der Kleij et al.

    A novel host-parasite lipid cross-talk. Schistosomal lyso-phosphatidylserine activates toll-like receptor 2 and affects immune polarization

    J. Biol. Chem

    (2002)
  • J.V. Weinstock et al.

    Translatability of helminth therapy in inflammatory bowel diseases

    Int. J. Parasitol

    (2013)
  • D.M.W. Zaiss et al.

    Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor

    Immunity

    (2013)
  • P. Adisakwattana et al.

    Helminth-derived immunomodulatory molecules

    Adv. Exp. Med. Biol

    (2009)
  • S. Amu et al.

    Regulatory B cells prevent and reverse allergic airway inflammation via FoxP3-positive T regulatory cells in a murine model

    J. Allergy Clin. Immunol

    (2010)
  • M.I. Araujo et al.

    Human schistosomiasis decreases immune responses to allergens and clinical manifestations of asthma

    Chem. Immunol. Allergy

    (2006)
  • A.M. Baru et al.

    Absence of Foxp3+ regulatory T cells during allergen provocation does not exacerbate murine allergic airway inflammation

    PLoS ONE

    (2012)
  • J. Bortolatto et al.

    Toll-like receptor 4 agonists adsorbed to aluminium hydroxide adjuvant attenuate ovalbumin-specific allergic airway disease: role of MyD88 adaptor molecule and interleukin-12/interferon-gamma axis

    Clin. Exp. Allergy

    (2008)
  • P.J. Cooper

    Interactions between helminth parasites and allergy

    Curr. Opin. Allergy Clin. Immunol

    (2009)
  • P.J. Cooper et al.

    Human onchocerciasis and tetanus vaccination: impact on the postvaccination antitetanus antibody response

    Infect. Immun

    (1999)
  • E. Danilowicz-Luebert et al.

    Modulation of specific and allergy-related immune responses by helminths

    J. Biomed. Biotechnol

    (2011)
  • C.N. Davis et al.

    MyD88-dependent and -independent signaling by IL-1 in neurons probed by bifunctional Toll/IL-1 receptor domain/BB-loop mimetics

    Proc. Natl. Acad. Sci. U.S.A.

    (2006)
  • F. Ebner et al.

    Therapeutic potential of larval excretory/secretory proteins of the pig whipworm Trichuris suis in allergic disease

    Allergy

    (2014)
  • S.C. Eisenbarth

    Use and limitations of alum-based models of allergy

    Clin. Exp. Allergy

    (2008)
  • S.C. Eisenbarth et al.

    Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants

    Nature

    (2008)
  • H. Evans et al.

    Worms as therapeutic agents for allergy and asthma: understanding why benefits in animal studies have not translated into clinical success

    J. Allergy Clin. Immunol

    (2014)
  • J.R. Feary et al.

    Experimental hookworm infection: a randomized placebo-controlled trial in asthma

    Clin. Exp. Allergy

    (2010)
  • C.M. Finlay et al.

    Induction of regulatory cells by helminth parasites: exploitation for the treatment of inflammatory diseases

    Immunol. Rev

    (2014)
  • L. Franchi et al.

    The Nlrp3 inflammasome is critical for aluminium hydroxide-mediated IL-1beta secretion but dispensable for adjuvant activity

    Eur. J. Immunol

    (2008)
  • A.L. Gavin et al.

    Adjuvant-enhanced antibody responses in the absence of toll-like receptor signaling

    Science

    (2006)
  • S. Ghosh et al.

    mu-chain-deficient mice possess B-1 cells and produce IgG and IgE, but not IgA, following systemic sensitization and inhalational challenge in a fungal asthma model

    J. Immunol

    (2012)
  • M.J. Gold et al.

    Group 2 innate lymphoid cells facilitate sensitization to local, but not systemic, TH2-inducing allergen exposures

    J. Allergy Clin. Immunol

    (2014)
  • Cited by (21)

    • IL-33: A central cytokine in helminth infections

      2021, Seminars in Immunology
      Citation Excerpt :

      Much of this work has been carried out using mouse models of asthma as a test-bed for type 2 immune response induction: this work led on from the seminal finding that H. polygyrus-infected mice were protected from allergic airway responses [133]. This effect could be replicated by administration of HES in the absence of infection [134], and correlated with reduced IL-33-dependent ILC2 activation [48,135]. Multiple pathways have been proposed or demonstrated for the IL-33-suppressive effects of HES, most recently with our identification of 2 families of IL-33-suppressive proteins secreted by the parasite [38,49,50,136,137] (Fig. 2).

    • TLR9 mediates the activation of NLRP3 inflammasome and oxidative stress in murine allergic airway inflammation

      2020, Molecular Immunology
      Citation Excerpt :

      It has been shown evidence of MyD88- and TRIF-independent TLR signaling in mice, and MyD88−/− mice develop normal airway inflammation in the OVA model as compared to wild-types (McSorley et al., 2015). Therefore, it is possible that OVA exposure may activate TLR9 signaling through unconventional signaling pathways(McSorley et al., 2015) and contribute to airway inflammation(H. M. Wu et al., 2016), while TLR9 stimulation with purified CpG-ODN protects from airway inflammation via MyD88 signaling and production of IL-10(Mirotti et al., 2017; Sabatel et al., 2017). Furthermore, in this study we further found that OVA challenge resulted in significant increase in the production of IL-5 and GM-CSF, both are well known mediators capable of inducing eosinophil differentiation, survival and migration (Asquith et al., 2008), however these two Th2 cytokines were substantially suppressed in OVA-challenged TLR9−/− mice.

    • HpARI Protein Secreted by a Helminth Parasite Suppresses Interleukin-33

      2017, Immunity
      Citation Excerpt :

      Many researchers, ourselves included, have demonstrated that helminths release immunomodulatory proteins to control anti-parasite immune responses and maintain their persistence in the host (Maizels and McSorley, 2016). We previously showed that the excretory–secretory products of the mouse intestinal parasite Heligmosomoides polygyrus (HES) suppress allergic responses in mouse models of asthma (Buck et al., 2014; McSorley et al., 2015; McSorley et al., 2014; McSorley et al., 2012). HES administration blocks the interleukin-33 (IL-33) response to inhaled Alternaria (fungal) allergen (McSorley et al., 2014) leading to reduced type 2 innate lymphoid cell (ILC2) responses and abrogating lung pathology.

    • The Role of Inflammasomes in Adjuvant-Driven Humoral and Cellular Immune Responses

      2017, Immunopotentiators in Modern Vaccines: Second Edition
    View all citing articles on Scopus
    View full text