ArticlePublisher preview available

Mast cell responses in a mouse model of food allergy are regulated via a ST2/IL‐4 axis

Wiley
Allergy
Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Figures

This content is subject to copyright. Terms and conditions apply.
   
|
2561
LETTER
5. Bart ko EA, Blom LH , Elberling J, Poul sen LK, Jensen B M. Expressio n
of CCR8 and CCX- CKR on basophils in chronic urticaria is ampli-
fied by IgE- mediated activation. Biomedicine. 2023;11(6):1537.
doi:10.3390/biomedicines11061537
6. Alhamwe BA, Potaczek DP, Miethe S, et al. Extracellular ves-
icles and asthma—more than just a co- existence. Int J Mol Sci.
2021;22(9):4984. doi:10.3390/ijms22094984
7. Botha J, Handberg A, Simonsen JB . Lipid- based str ategies used to
identif y extracellular vesicles in flow c ytometry can be confounded
by lipoproteins: evaluations of annexin V, lactadherin, and deter-
gent lysis. J Extracell Vesicles. 2022;11(4):e12200. doi:10.1002/
jev2.12200
SUPPORTING INFORMATION
Additional supporting information can be found online in the
Suppor ting Information section at the end of this article.
DOI : 10.1111/all.162 57
Mast cell responses in a mouse model of food allergy are
regulated via a ST2/IL- 4 axis
To the Editor,
Allergen- induced mast cell (MC) activation is critical for the de-
velopment of food allergy. Recent evidence implicates the alarmin
cytokine, IL- 33, as a key player in regulating MC responses dur-
ing allergic inflammation. The IL- 33 receptor, ST2, is constitutively
expressed on MCs, and genetic polymorphisms within the IL- 33/
ST2 axis are strongly linked to disease susceptibility.1 Several re-
cent studies have demonstrated a critical role for IL- 33 in inducing
MC responses to food antigens, including decreased MC activation
and food allergy development in both epicutaneously- sensitized
ST2−/− mice2,3 and mice treated with anti- IL- 33 antibodies.2,4
Similarly, IL- 33- stimulated group 2 innate lymphoid cells can also
activate MCs in enterally- challenged mice.5 However, depend-
ing on the model of allergic sensitization (epicutaneous—with or
without t ape- stripping), divergent effects on MC activation, in-
testinal MC numbers, and anaphylaxis have been observed.2, 3
While epicutaneously- sensitized, tape- stripped ST2−/− mice were
protected from anaphylaxis due to decreased MC activation, no
changes in intestinal MC expansion were observed.2 In contrast , in
non- tape- stripped ST2−/− mice, both MC activation and intestinal
MC numbers were decreased.3
We sought to investigate the role of IL- 33 using a well-
established model of systemic sensitization to food antigens. Briefly,
wild- type Balb/c (WT) and ST2−/− mice were immunized i.p. with
chicken egg ovalbumin (OVA), followed by intragastric OVA chal-
lenges. Oral allergen exposure resulted in profuse allergic diarrhea
in sensitized WT mice (Figure 1A), accompanied by a robust antigen-
mediated IgE response (Figure 1B), significant MC accumulation in
the jejunum (Figure 1C), and increased MC activation (Figure 1D),
as indicated by elevated serum MC protease- 1 (MCPT- 1). In con-
trast, ST2−/− mice exhibited decreased diarrhea and MC responses
(Figure 1A–D). Similarly, the expression of the t ype 2 cytokine
genes, IL- 4 and IL- 13, was also reduced in the intestines of ST2−/−
mice (Figure 1E,F). In contrast, enhanced expression of IL- 33 was
observed in OVA- sensitized ST2−/− mice (Figure S1 A). A trend to-
wards increased expression of IFN- γ was also obser ved (Figure S1
B), suggesting that a decrease in type 2 cytokines may be accom-
panied by a parallel increase in type 1 cytokines. IL- 33 is critical for
the induction of allergen- specific Th2 cells.6,7 We therefore won-
dered whether systemic T cell responses may also be impaired in
the absence of ST2. As obser ved in Figure 1G–J, while splenocytes
from OVA- challenged WT mice secreted elevated levels of IL- 4, IL- 5
and IL- 13 in response to antigen restimulation, the production of
these cytokines was decreased in the ST2−/− group. Similarly, T cell
receptor- mediated polyclonally- induced type 2 cytokines were also
decreased in OVA- challenged ST2−/− mice (Figure S1 C–F ).
Considering the surprisingly low levels of T cell- derived IL- 4 in
the absence of IL- 33 signa ling and given that IL- 4 has been shown to
induce MC activation,8 we wondered whether the attenuated MC
responses in OVA- sensitized ST2−/− mice may be a consequence of
decreased IL- 4 produc tion. This is consistent with previous stud-
ies demonstrating decreased IL- 4 production in antigen- specific T
cells from ST2−/− mice.9 Furthermore, while we did not observe any
differences in baseline IL- 4 production from polyclonally stimu-
lated naïve WT or ST2−/− splenic T cells (Figure S2A and as has also
been observed previously9), IL- 4Rα expression was decreased in
both bone marrow- derived MCs (Figure S2B) and OVA- sensitized
CD4 T cells (Figure S2C) from ST2−/− mice. Lastly, the induction
of food allergy in this model has also been shown to be depen-
dent on IL- 4 with both antibody- mediated depletion and genetic
ablation resulting in the suppression of MC responses.10 To there-
fore further assess the role of IL- 4, we treated mice with IL- 4C to
boost IL- 4 levels in ST2−/− mice. To our surprise, IL- 4 treatment not
only amplified the effec ts of allergen challenge in W T animals, but
© 2024 EA ACI and John Wiley and Sons A /S. Published by John Wiley and Sons Ltd.
... In a similar vein, we recently also demonstrated a critical role for IL-33 in inducing MC expansion and activation during food allergy development (26). IL-33 is a potent stimulator of MCs and can promote both IgE-dependent and independent MC-mediated inflammation (27)(28)(29)(30)(31)(32). ...
... Allergic responses to enteral ovalbumin administration are suppressed in ST2 -/mice Recently, IL-33 has emerged as a critical mediator of MC responses during food allergy (8,26,(49)(50)(51). To further investigate the crosstalk between IL-10 and IL-33, and whether IL-10's effects on IgE-induced MC responses may depend on IL-33, we assessed the development of food allergy in WT and IL-10depleted ST2 -/mice using an ovalbumin (OVA)-induced model of intestinal anaphylaxis. ...
... We and others have previously shown that the development of food allergy in this model is IgE and MCdependent (9,14,43,46,52). Similarly, we recently also demonstrated a critical role for IL-33 in inducing MC responses in this model (26). Lastly, using both IL-10 -/mice (14) as well as pharmacological blockade of IL-10 (17), we have shown that MC responses and the development of intestinal anaphylaxis in this model are also IL-10-dependent. ...
Article
Full-text available
Background The IL-33/ST2 axis plays a pivotal role in the development of IgE-mediated mast cell (MC) responses during food allergy. We recently demonstrated that the pleiotropic cytokine, IL-10, not only exerts proinflammatory effects on IgE-mediated MC activation, but also promotes IL-33-induced MC responses. However, whether IL-33 is necessary for IL-10’s proinflammatory effects has not been examined. Methods To therefore determine the role of the IL-33/ST2 axis in this pathway, we assessed the effects of IL-10 on IgE-mediated MC activation and food allergy development in wild-type (WT) and ST2-/- mice. Results IL-10 stimulation significantly enhanced IL-33 gene expression, ST2 receptor expression, cytokine production, mMCP-1 secretion, and proliferation in IgE and antigen-activated bone marrow-derived MCs (BMMCs) from WT mice. ST2-/- BMMCs exhibited reduced cytokine secretion in response to IgE-dependent activation. However, IL-10 enhanced cytokine production, mMCP-1 secretion, and proliferation in these cells as well. To further assess the role of IL-10, food allergy was induced in WT and ST2-/- mice subjected to antibody-mediated IL-10 depletion. IL-10-depleted WT mice exhibited a significant attenuation in MC-mediated responses to OVA challenge. While ST2-/- mice also exhibited a profound suppression of MC responses, IL-10 depletion had no additional effects. However, ST2-/-/IL-10-/- mice exhibited further decreases in OVA-IgE and antigen-specific MC activation compared to ST2-/- mice. Conclusion Our data demonstrates that IL-10 can enhance MC responses in both WT and ST2-/- mice, further corroborating its proinflammatory effects on MCs and suggesting that they are not regulated by IL-33 signaling.
Article
Full-text available
Type 2‐mediated immune responses protect the body against environmental threats at barrier surfaces, such as large parasites and environmental toxins, and facilitate the repair of inflammatory tissue damage. However, maladaptive responses to typically nonpathogenic substances, commonly known as allergens, can lead to the development of allergic diseases. Type 2 immunity involves a series of prototype TH2 cytokines (IL‐4, IL‐5, IL‐13) and alarmins (IL‐33, TSLP) that promote the generation of adaptive CD4+ helper Type 2 cells and humoral products such as allergen‐specific IgE. Mast cells and basophils are integral players in this network, serving as primary effectors of IgE‐mediated responses. These cells bind IgE via high‐affinity IgE receptors (FcεRI) expressed on their surface and, upon activation by allergens, release a variety of mediators that regulate tissue responses, attract and modulate other inflammatory cells, and contribute to tissue repair. Here, we review the biology and effector mechanisms of these cells, focusing primarily on their role in mediating IgE responses in both physiological and pathological contexts.
Article
Full-text available
BACKGROUNDIL-33, found in high levels in participants with allergic disorders, is thought to mediate allergic reactions. Etokimab, an anti-IL-33 biologic, has previously demonstrated a good safety profile and favorable pharmacodynamic properties in many clinical studies.METHODS In this 6-week placebo-controlled phase 2a study, we evaluated the safety and the ability of a single dose of etokimab to desensitize peanut-allergic adults. Participants received either etokimab (n = 15) or blinded placebo (n = 5). Clinical tests included oral food challenges and skin prick tests at days 15 and 45. Blood samples were collected for IgE levels and measurement of ex vivo peanut-stimulated T cell cytokine production.RESULTSEfficacy measurements for active vs. placebo participants at the day 15 and 45 food challenge (tolerating a cumulative 275 mg of peanut protein, which was the food challenge outcome defined in this paper) demonstrated, respectively, 73% vs. 0% (P = 0.008) to 57% vs. 0% (ns). The etokimab group had fewer adverse events compared with placebo. IL-4, IL-5, IL-9, IL-13, and ST2 levels in CD4+ T cells were reduced in the active vs. placebo arm upon peanut-induced T cell activation (P = 0.036 for IL-13 and IL-9 at day 15), and peanut-specific IgE was reduced in active vs. placebo (P = 0.014 at day 15).CONCLUSION The phase 2a results suggest etokimab is safe and well tolerated and that a single dose of etokimab could have the potential to desensitize peanut-allergic participants and possibly reduce atopy-related adverse events.TRIAL REGISTRATIONClinicalTrials.gov NCT02920021.FUNDINGThis work was supported by NIH grant R01AI140134, AnaptysBio, the Hartman Vaccine Fund, and the Sean N. Parker Center for Allergy and Asthma Research at Stanford University.
Article
Full-text available
Interleukin-33 (IL-33) is a tissue-derived nuclear cytokine from the IL-1 family abundantly expressed in endothelial cells, epithelial cells and fibroblast-like cells, both during homeostasis and inflammation. It functions as an alarm signal (alarmin) released upon cell injury or tissue damage to alert immune cells expressing the ST2 receptor (IL-1RL1). The major targets of IL-33 in vivo are tissue-resident immune cells such as mast cells, group 2 innate lymphoid cells (ILC2s) and regulatory T cells (Tregs). Other cellular targets include T helper 2 (Th2) cells, eosinophils, basophils, dendritic cells, Th1 cells, CD8⁺ T cells, NK cells, iNKT cells, B cells, neutrophils and macrophages. IL-33 is thus emerging as a crucial immune modulator with pleiotropic activities in type-2, type-1 and regulatory immune responses, and important roles in allergic, fibrotic, infectious, and chronic inflammatory diseases. The critical function of IL-33/ST2 signaling in allergic inflammation is illustrated by the fact that IL33 and IL1RL1 are among the most highly replicated susceptibility loci for asthma. In this review, we highlight 15 years of discoveries on IL-33 protein, including its molecular characteristics, nuclear localization, bioactive forms, cellular sources, mechanisms of release and regulation by proteases. Importantly, we emphasize data that have been validated using IL-33-deficient cells.
Article
Full-text available
Background Food allergy (FA) is an increasing problem that has no approved treatment. The pro-Th2 cytokines, IL-25, IL-33 and TSLP, are associated with FA and monoclonal antibodies (mAbs) to these cytokines are reported to suppress murine FA development. Objective Determine whether anti-pro-Th2 cytokine mAbs can block both FA maintenance and induction. Methods IgE-mediated FA was induced in BALB/c mice by oral gavage (o.g.) with medium chain triglycerides plus egg white (MCT/EW) and was characterized by increased numbers of lamina propria Th2 cells, mast cells shock, and eosinophils, shock (hypothermia), mast cell degranulation (increased serum MMCP1), increased serum IgG1 anti-EW and IgE, and increased IL-4 and IL-13 secretion following MCT/EW challenge. To suppress FA development, mice were injected with anti-IL-25, IL-33R, and/or TSLP monoclonal antibodies prior to the initial o. g. with MCT/EW; to suppress established FA, treatment with the same mAbs was initiated after FA development. Results Injection of a mAb to IL-25, IL-33R, or TSLP strongly inhibited FA development. No single mAb to a pro-Th2 cytokine could suppress established FA and optimal FA suppression required treatment with a cocktail of all three anti-pro-Th2 mAbs. Treatment with the three mAb cocktail during initial MCT/EW immunization induced EW tolerance. Conclusion All of the pro-Th2 cytokines are required to induce our model of FA, while any pro-Th2 cytokine can maintain established FA. Pro-Th2 cytokines prevent oral tolerance. Combined treatment with antagonists to all three pro-Th2 cytokines or with an inhibitor of pro-Th2 cytokine production may be able to suppress established human FA.
Article
Full-text available
Background: Cutaneous exposure to food allergens predisposes to food allergy, which is commonly associated with atopic dermatitis (AD). Levels of the epithelial cytokine IL-33 are increased in skin lesions and serum of patients with AD. Mast cells (MCs) play a critical role in food-induced anaphylaxis and express the IL-33 receptor ST2. The role of IL-33 in patients with MC-dependent food anaphylaxis is unknown. Objective: We sought to determine the role and mechanism of action of IL-33 in patients with food-induced anaphylaxis in a model of IgE-dependent food anaphylaxis elicited by oral challenge of epicutaneously sensitized mice. Methods: Wild-type, ST2-deficient, and MC-deficient Kit(W-sh/W-sh) mice were epicutaneously sensitized with ovalbumin (OVA) and then challenged orally with OVA. Body temperature was measured by means of telemetry, Il33 mRNA by means of quantitative PCR, and IL-33, OVA-specific IgE, and mouse mast cell protease 1 by means of ELISA. Bone marrow-derived mast cell (BMMC) degranulation was assessed by using flow cytometry. Results: Il33 mRNA expression was upregulated in tape-stripped mouse skin and scratched human skin. Tape stripping caused local and systemic IL-33 release in mice. ST2 deficiency, as well as ST2 blockade before oral challenge, significantly reduced the severity of oral anaphylaxis without affecting the systemic TH2 response to the allergen. Oral anaphylaxis was abrogated in Kit(W-sh/W-sh) mice and restored by means of reconstitution with wild-type but not ST2-deficient BMMCs. IL-33 significantly enhanced IgE-mediated degranulation of BMMCs in vitro. Conclusion: IL-33 is released after mechanical skin injury, enhances IgE-mediated MC degranulation, and promotes oral anaphylaxis after epicutaneous sensitization by targeting MCs. IL-33 neutralization might be useful in treating food-induced anaphylaxis in patients with AD.
Article
Full-text available
Background: Food allergy is a major health issue, but its pathogenesis remains obscure. Group 2 innate lymphoid cells (ILC2s) promote allergic inflammation. However their role in food allergy is largely unknown. Objective: We sought to investigate the role of ILC2s in food allergy. Methods: Food allergy-prone mice with a gain-of-function mutation in the IL-4 receptor α chain (Il4raF709) were orally sensitized with food allergens, and the ILC2 compartment was analyzed. The requirement for ILC2s in food allergy was investigated by using Il4raF709, IL-33 receptor-deficient (Il1rl1(-/-)), IL-13-deficient (Il13(-/-)), and IL-4-deficient (Il4(-/-)) mice and by adoptive transfer of in vitro-expanded ILC2s. Direct effects of ILC2s on regulatory T (Treg) cells and mast cells were analyzed in coculture experiments. Treg cell control of ILC2s was assessed in vitro and in vivo. Results: Il4raF709 mice with food allergy exhibit increased numbers of ILC2s. IL-4 secretion by ILC2s contributes to the allergic response by reducing allergen-specific Treg cell and activating mast cell counts. IL-33 receptor deficiency in Il4raF709 Il1rl1(-/-) mice protects against allergen sensitization and anaphylaxis while reducing ILC2 induction. Adoptive transfer of wild-type and Il13(-/-) but not Il4(-/-) ILC2s restored sensitization in Il4raF709 Il1rl1(-/-) mice. Treg cells suppress ILC2s in vitro and in vivo. Conclusion: IL-4 production by IL-33-stimulated ILC2s blocks the generation of allergen-specific Treg cells and favors food allergy. Strategies to block ILC2 activation or the IL-33/IL-33 receptor pathway can lead to innovative therapies in the treatment of food allergy.
Article
Full-text available
T helper (Th) cells can be categorized according to their cytokine expression. The differential induction of Th cells expressing Th1 and/or Th2 cytokines is key to the regulation of both protective and pathological immune responses. Cytokines are expressed transiently and there is a lack of stably expressed surface molecules, significant for functionally different types of Th cells. Such molecules are of utmost importance for the analysis and selective functional modulation of Th subsets and will provide new therapeutic strategies for the treatment of allergic or autoimmune diseases. To this end, we have identified potential target genes preferentially expressed in Th2 cells, expressing interleukin (IL)-4, IL-5, and/or IL-10, but not interferon-γ. One such gene, T1/ST2, is expressed stably on both Th2 clones and Th2-polarized cells activated in vivo or in vitro. T1/ST2 expression is independent of induction by IL-4, IL-5, or IL-10. T1/ST2 plays a critical role in Th2 effector function. Administration of either a mAb against T1/ST2 or recombinant T1/ST2 fusion protein attenuates eosinophilic inflammation of the airways and suppresses IL-4 and IL-5 production in vivo following adoptive transfer of Th2 cells.
Article
Full-text available
Interleukin (IL)-4 has critical roles in allergic disorders, including food hypersensitivity. The direct effects of the cytokine on the survival and function of mast cells, the key effectors of food anaphylaxis, have not been established. In this study, we demonstrate that IL-4 induces a marked intestinal mastocytosis in mice. This phenotype is reproduced in animals expressing Il4rαF709, an activating variant of the IL-4 receptor α-chain (IL-4Rα). Il4rαF709 mice exhibit enhanced anaphylactic reactions but unaltered physiological responses to vasoactive mediators. IL-4 induces Bcl-2 and Bcl-X(L) and enhances survival and stimulates proliferation in cultured bone marrow-derived mast cells (BMMC). These effects are STAT6 (signal transducer and activator of transcription factor 6)-dependent and are amplified in Il4rαF709 BMMC. In competitive bone marrow chimeras, Il4rαF709 mast cells display a substantial competitive advantage over wild-type mast cells, which, in turn, prevail over IL-4Rα(-/-) mast cells in populating the intestine, establishing a cell-intrinsic effect of IL-4 in intestinal mast cell homeostasis. Our results demonstrate that IL-4-signaling is a key determinant of mast cell expansion in food allergy.Mucosal Immunology advance online publication 14 November 2012; doi:10.1038/mi.2012.112.
Article
Full-text available
We have generated mice with a deficiency in T1/ST2 expression to clarify the roles of T1/ST2 in T helper cell type 2 (Th2) responses. Using immunological challenges normally characterized by a Th2-like response, we have compared the responses of T1/ST2-deficient mice with those generated by wild-type mice. Using a primary pulmonary granuloma model, induced with Schistosoma mansoni eggs, we demonstrate that granuloma formation, characterized by eosinophil infiltration, is abrogated in T1/ST2-deficient mice. Furthermore, we clearly demonstrate that in the absence of T1/ST2 expression, the levels of Th2 cytokine production are severely impaired after immunization. Thus, in a secondary pulmonary granuloma model, draining lymph node cells from the T1/ST2-deficient animals produced significantly reduced levels of IL-4 and IL-5, despite developing granulomas of a magnitude similar to those of wild-type mice and comparable antigen-specific immunoglobulin isotype production. These data clearly demonstrate that T1/ST2 expression plays a role in the development of Th2-like cytokine responses and indicate that effector functions are inhibited in its absence.
Article
Background: A major route of sensitization to food allergen is through an impaired skin barrier. IL33 and TSLP have both been implicated in epicutaneous sensitization and food allergy, albeit in different murine models. Objective: We assessed the respective contributions of TSLP and IL33 to the development of atopic dermatitis (AD) and subsequent food allergy in TSLP and IL33 receptor (ST2) deficient mice using an AD model that does not require tape stripping. Method: TSLPR-/-, ST2-/- and BALB/cJ control mice were exposed to 3 weekly epicutaneous skin patches of either saline, OVA, or a combination of OVA and Aspergillus fumigatus (ASP), followed by repeated intragastric OVA challenges and development of food allergy. Results: ASP(+/-OVA)-patched, but not OVA-patched, BALB/cJ mice developed an AD-like skin phenotype. However, epicutaneous OVA sensitization occurred in OVA-patched mice and was decreased in ST2-/- mice resulting in lower intestinal mast cell degranulation, accumulation and OVA-induced diarrhea occurrences upon intragastric OVA challenges. In TSLPR-/- mice, intestinal MC accumulation was abrogated, and no diarrhea was observed. AD was significantly milder in OVA+ASP-patched TSLPR-/- mice compared to wild type and ST2-/- mice. Accordingly, intestinal mast cell accumulation and degranulation were impaired in OVA+ASP-patched TSLPR-/- mice compared to wild type and ST2-/- mice, protecting TSLPR-/- mice from developing allergic diarrhea. Conclusion: Epicutaneous sensitization to food allergen and development of food allergy can occur without skin inflammation and is partly mediated by TSLP, suggesting that prophylactic targeting of TSLP may be useful in mitigating the development of atopic dermatitis and food allergy early in life in at-risk infants.
Article
Intestinal anaphylaxis (manifested by acute diarrhea) is dependent on IgE and mast cells. We aimed to define the respective roles of IL-4 and IL-13 and their receptors in disease pathogenesis. Wild-type mice and mice deficient in IL-4, IL-13, and IL-13 receptor (IL-13R) alpha1 (part of the type 2 IL-4 receptor [IL-4R]) were sensitized with ovalbumin (OVA)/aluminum potassium sulfate and subsequently given repeated intragastric OVA exposures. The IL-4R alpha chain was targeted with anti-IL-4R alpha mAb before or after intragastric OVA exposures. IL4(-/-) (and IL4/IL13(-/-)) mice produced almost no IgE and were highly resistant to OVA-induced diarrhea, whereas allergic diarrhea was only partially impaired in IL13(-/-) and IL13Ralpha1(-/-) mice. IL13Ralpha1-deficient mice had decreased IgE levels, despite having normal baseline IL-4 levels. Intestinal mast cell accumulation and activation also depended mainly on IL-4 and, to a lesser extent, on IL-13. Prophylactic anti-IL-4R alpha mAb treatment, which blocks all IL-4 and IL-13 signaling, suppressed development of allergic diarrhea. However, treatment with anti-IL-4R alpha mAb for 7 days only partially suppressed IgE and did not prevent intestinal diarrhea. Endogenously produced IL-13 supplements the ability of IL-4 to induce allergic diarrhea by promoting oral allergen sensitization rather than the effector phase of intestinal anaphylaxis.