Food allergies represent an increasingly prevalent human health problem, and therapeutic options remain limited, with avoidance being mainstay, despite its adverse effects on quality of life. A better understanding of the key immunological mechanisms involved in such responses likely will be vital for development of new therapies. This review outlines the current understanding of how the immune system is thought to contribute to prevention or development of food allergies. Drawing from animal studies, as well as clinical data when available, the importance of oral tolerance in sustaining immunological nonresponsiveness to food Ags, our current understanding of why oral tolerance may fail and sensitization may occur, and the knowledge of pathways that may lead to anaphylaxis and food allergy–associated responses are addressed.

Within the clinical realm of allergy, food allergy is receiving an increasing amount of attention, mirroring its increasing prevalence both nationally and internationally. Current estimates put food allergy as affecting up to 15 million people within the United States (1). Therapeutically, these patients are dependent on a difficult avoidance approach, with injectable epinephrine as a life-saving option in case of accidental exposure. This has been shown to significantly affect quality of life (2), and recent advances in understanding the mechanisms behind food allergy have been fueled by the desire to develop improved therapies.

In considering such mechanisms, we focus on three processes that may be important: oral tolerance, sensitization to food allergens, and anaphylactic reactivity to these food allergens. Finally, an emerging concept of “nonresponsive tolerance,” where anaphylactic reactivity does not occur or is lost despite evidence for IgE-associated sensitization is highlighted.

Oral tolerance to egg proteins was first described >100 y ago (3). This natural phenomenon, where ingested food proteins do not elicit a specific immune response, is also observed in humans (4), but the necessary mechanisms remain unclear. Despite gastrointestinal enzymes degrading food and the physical barrier of the intestinal mucosa, immune surveillance of food Ags and establishment of tolerance mechanisms are clearly occurring. Several reviews addressed possible routes of Ag sampling and presentation (57), including sampling by dendritic cells (DCs) across the epithelial layer, presentation by M cells or goblet cells to DCs, or soluble Ag directly traversing the epithelium through paracellular or transcellular routes.

Key cells seem important for oral tolerance and the maintenance of regulatory (Foxp3+) T cell (Treg) populations (Fig. 1). CD11cCD11b+F4/80+ macrophages exhibit an anti-inflammatory gene signature and produce IL-10 (8). Additionally, two distinct subsets of tolerance-associated CD11c+ cells reside in the intestinal lamina propria, expressing either CX3CR1 or CD103 (9). CX3CR1 knockout mice show diminished IL-10 production and Treg populations, as well as a lack of oral tolerance in a food allergy model (10). In contrast, CX3CR1+CD103 cells have been implicated in intestinal inflammation (11).

FIGURE 1.

Within the intestine, unique populations of cells that include macrophages, CX3CR1+ APCs, or CD103+ DCs ensure maintenance of tolerance through driving development of IL-10–producing Tregs and IgA-secreting B cells. Critical signals for tolerance are provided by retinoic acid (RA), IDO, and TGF-β. Perturbation in these cells or mediators, through largely unknown signals, breaks tolerance and promotes allergic sensitization characterized by dominant Th2-biased responses and class-switching toward IgG and IgE. Evidence supports the roles of tissue-derived cytokines, particularly IL-33, in supporting these events, perhaps via activation of innate lymphoid cells (ILCs). Initiating signals for sensitization include intrinsic activities of food components on innate cells, such as NKT cells, and exposure to bacterial toxins, such as SEB. The intestinal microbiota also may influence the balance between tolerance and sensitization. Additionally, defective barrier functions at either the skin or intestine were shown to facilitate sensitization to food allergens.

FIGURE 1.

Within the intestine, unique populations of cells that include macrophages, CX3CR1+ APCs, or CD103+ DCs ensure maintenance of tolerance through driving development of IL-10–producing Tregs and IgA-secreting B cells. Critical signals for tolerance are provided by retinoic acid (RA), IDO, and TGF-β. Perturbation in these cells or mediators, through largely unknown signals, breaks tolerance and promotes allergic sensitization characterized by dominant Th2-biased responses and class-switching toward IgG and IgE. Evidence supports the roles of tissue-derived cytokines, particularly IL-33, in supporting these events, perhaps via activation of innate lymphoid cells (ILCs). Initiating signals for sensitization include intrinsic activities of food components on innate cells, such as NKT cells, and exposure to bacterial toxins, such as SEB. The intestinal microbiota also may influence the balance between tolerance and sensitization. Additionally, defective barrier functions at either the skin or intestine were shown to facilitate sensitization to food allergens.

Close modal

Most evidence supports a role for CX3CR1CD103+ DCs in tolerance. These cells exhibit lymph-node homing where they activate naive T cells (9, 12) and promote a FoxP3+ Treg phenotype, a process requiring both TGF-β and retinoic acid (1315). Retinoic acid imprints the gut-homing receptors CCR9 and α4β7 onto both Tregs (16) and IgA-secreting B cells (17), an event that also seems to contribute to oral tolerance (16). CD103+ DCs also use IDO for tolerance, and loss of IDO function drives T cells toward a Th1 or Th17 phenotype, limiting Tregs and oral tolerance (18). Recent findings also show that MUC2, a mucin secreted by intestinal goblet cells, supports the anti-inflammatory potential of these CD103+ DCs (19).

IPEX (immune dysregulation, polyendocrinopathy, enteropathy, X-linked) patients with mutations in the FOXP3 locus (20) develop severe food allergy, as well as a plethora of other disorders, including autoimmunity, enteropathy, and atopic dermatitis (21), indicating the importance of Tregs in tolerance. Foxp3-mutant mice (scurfy) and DEREG mice, in which Tregs can be deleted upon diphtheria toxin treatment, also have been used to demonstrate the importance of Tregs in allergic responses (22, 23). Expression of CCR9 and α4β7 on Tregs is necessary for tolerance, because these molecules support gut homing (10, 16). Although previous reviews summarized the effects of Ag concentration in oral tolerance (5, 6) [i.e., low doses drive Tregs, whereas high doses yield anergy and deletion of T cells (24)], most evidence points toward Treg-associated low-dose tolerance as being critical in food allergy. We (25) showed previously that loss of oral tolerance to peanut was associated with diminished Treg responses but also that high-dose Ag feeding could overcome allergic responses.

In food allergy, the immune response is clearly biased toward a type 2 cytokine–associated phenotype. Why specific food Ags trigger this response remains unclear, although some food Ags possess the potential to stimulate innate immune responses. For example, the peanut allergen Ara h1 binds to CD209 on DCs (26), and milk sphingomyelin activates type 2 cytokine responses from invariant NKT cells (27).

Changes in microbial flora have been associated with allergic sensitization, with several lines of evidence supporting protection by specific bacteria and their products, likely through sustaining intestinal Treg populations (reviewed in Ref. 28). Mice with decreased commensal bacteria colonies, which includes either germ-free or antibiotic-treated mice, exhibit increased food-allergen sensitivity (29), high serum IgE, and increased circulating basophils (30). Interestingly, mice with enhanced signaling through IL-4Rα who display profound allergic sensitization and anaphylaxis to food Ags (31) also exhibit an altered microbiota that can be normalized by Treg transfer (32). Consequently, the interaction of host immunity and commensal microbiota seems bidirectional, with host immune responses not just responding to bacterial presence but also shaping the bacterial flora toward that associated with pathology.

The critical mechanisms responsible for allergic sensitization are beginning to be elucidated. Generally in allergy, epithelial production of TSLP, IL-25, and IL-33 has become a key area of interest (33). However, a recent study (34) of these cytokines using a cholera toxin–driven oral peanut model showed that only IL-33 was required for sensitization. IL-33 can increase mucosal permeability (35) and promote Th2 skewing by DCs (36). Interestingly, although constitutive IL-33 expression occurs in epithelial cells, increasing evidence supports the potential for inducible expression by several immune cells, including DCs, that is sufficient for subsequent Th2 immunity, as was shown in helminth infection and for IgG immune complexes (37, 38). However, the key producer of IL-33 in food allergy remains to be determined.

At the level of Ag presentation, several mechanisms that may participate in tipping the balance from tolerance to sensitization have been described. Binding of OX40 ligand to OX40, TIM4 to TIM1, and jagged to notch on DCs and naive T cells, respectively, can regulate T cell differentiation from Tregs toward Th2, as previously reviewed (39). Environmental interactions may drive this differentiation; for example, staphylococcal enterotoxin B (SEB) can break tolerance and promote food allergy (25, 40), and it was shown mechanistically to induce TIM4 expression on DCs that is necessary for Th2 skewing (40). Th2-associated responses also can occur if Tregs are deleted (10) or become dysfunctional, as induced by SEB (41). In contrast, some innate signals also may protect against sensitization, because TLR9−/− mice have impaired IgE and IgA responses, resulting in reduced anaphylaxis to peanut (42).

Intestinal penetrance by allergens also may enhance allergic sensitization (43). On intestinal epithelial cells, IL-4 can induce upregulation of the low-affinity IgE receptor, CD23, which binds Ag-specific IgE and facilitates Ag uptake (44). This potential mechanism may explain why large or low-solubility Ags traverse the epithelium and elicit systemic responses (45). Similarly, alterations in tight-junction integrity may allow Ag penetrance. For example, deficiency in the desmosomal ICAM desmoglein-1 was shown to elicit profound allergic responses (46), whereas its expression is reduced in tissues of patients with eosinophilic esophagitis (47).

Recent interest also has focused on the skin as a potential route for sensitization, because food allergy often associates with eczema in patients (1). Barrier integrity also may be important here, because filaggrin-deficient mice, which exhibit weak epithelial barrier function, become sensitized to proteins on the skin (48), and epicutaneous sensitization is sufficient to promote anaphylaxis upon oral challenge (49). Although very few studies have defined specific genes associated with food allergy, it is interesting to note that mutations in desmoglein-1 (47), filaggrin (50), and TSLP (51) were shown to be associated with food allergy or eosinophilic esophagitis in human cohorts, because these molecules all regulate skin homeostasis. However, it is unclear whether these associations relate to food allergy or eczema, because these diseases are often coincident in children, and the number of genes associated with food allergy alone remains relatively limited (52).

The mechanisms of anaphylaxis––the hallmark of food-allergy reactivity––are generally biphasic: an acute reaction occurs immediately after allergen exposure, followed by a late-phase reaction several hours later. Symptoms occurring during the acute reaction are due to release of preformed mediators, whereas the late-phase response involves influx of inflammatory cells. Clinically, heterogeneity in responses is observed, with some patients experiencing either the acute or late-phase reaction and others experiencing both the acute and late-phase reactions (53). In addition to clinical heterogeneity, anaphylactic responses can be elicited through multiple mechanisms.

First shown in 1997 by Miyajima et al. (54), both IgE and IgG can play a role in anaphylaxis in the mouse. IgE functions via its high-affinity receptor, FcεRI, which is highly expressed on mast cells and basophils (55). FcεRI−/− mice do not respond in a passive IgE-mediated systemic anaphylaxis model (56) and have reduced responses in models of allergic diarrhea and food allergy (5759). IgG has several receptors: the high-affinity FcγRI and FcγRIV and the low-affinity FcγRIIB and FcγRIII. All of these receptors are expressed on several cell types involved in anaphylaxis, including mast cells, basophils, neutrophils, and macrophages. Using a model of systemic anaphylaxis, Strait et al. (56) showed that inhibition of FcγRII/III abolished temperature drops associated with shock in IgG-mediated, but not IgE-mediated, anaphylaxis. Similarly, Jönsson et al. (60) used knockout mice to show that FcγRIV is necessary for systemic anaphylaxis. Although these pathways have been differentially defined using these passive models, both Abs appear to participate in active food allergy: Arias et al. (61) showed that IgE−/− and IgG1−/− mice were only partially protected from peanut-induced anaphylaxis, but blockade of IgG1 in IgE−/− mice completely abolished the response; similarly, FcRγ−/− mice, which lack the common chain for both the IgE and IgG receptors, were protected (62). Importantly, recent studies using humanized mice supported the potential anaphylactic functions of IgG via human receptors (63).

Histamine, platelet-activating factor (PAF), and 5-hydroxytryptamine (5-HT; serotonin) are all sufficient to induce early-phase anaphylaxis (64, 65). Several groups also looked at the necessity for each of these mediators in anaphylaxis, and there appears to be heterogeneity here also.

Histamine, produced from both mast cells and basophils, is a well-established mediator necessary for anaphylaxis (56, 66). In IgE-mediated systemic anaphylaxis, histamine synthesis, as well as histamine H1 and H2 receptors, is necessary for responses (66, 67), and blockade of these receptors is therapeutically beneficial in patients with acute allergic reactions (68).

Additionally, PAF and 5-HT were shown to contribute to anaphylaxis (56, 58, 6163). Several inflammatory cells make PAF, including macrophages/monocytes, mast cells, basophils, neutrophils, and platelets. Although associated with platelet activation, PAF also influences vascular permeability, leukocyte recruitment, and leukocyte activation (69). Studies using models of allergic diarrhea, food allergy, or systemic anaphylaxis models showed that responses may be due to either PAF and histamine (56, 61) or PAF and 5-HT (58, 63).

Although other mast cell– and basophil-derived mediators have been implicated in food allergy, their role is less defined. These include other preformed mediators (e.g., tryptase, chymase, and heparin), lipid mediators [e.g., PGD2, LTC4, LTD4, and LTE4 (70)], and several cytokines. IgE activation of mast cells has the potential to generate several cytokines that were shown to direct late-phase inflammation, including release of preformed TNF and synthesis of IL-33 (71, 72). TNF was shown to be necessary for late-phase recruitment of neutrophils (71), as well as for a late-phase increase in PAF in the serum (73). The IL-33 receptor, ST2, is necessary for IgE-triggered tissue inflammation (72). IL-33 does not directly cause mast cell degranulation (74), but it promotes expression of several cytokines and chemokines, including IL-6 and IL-13, from mast cells and eosinophils (72, 75). Similarly, IL-9 can both stimulate and be produced by mast cells (76). IL-9 was shown to be critical for the initiation and severity of food-associated anaphylaxis by promoting intestinal mastocytosis (77, 78).

Largely from murine studies of passive sensitization models, mast cells, basophils, macrophages, and neutrophils were shown to contribute to anaphylactic shock responses. Four distinct pathways of response seem to be possible: a “classic” pathway involving IgE, FcεRI, mast cells, and histamine; an “alternative” pathway mediated by IgG1, FcγRIII, macrophages, and PAF (79); an IgG–basophil–PAF pathway (80); and an IgG–neutrophil–PAF pathway via FcγRIV activation (Fig. 2) (60).

FIGURE 2.

Multiple pathways of anaphylaxis exist, mediated by either IgE or IgG and their respective FcRs. Within tissues, mast cell activation via IgE and FcεRI initiates early-phase responses mediated by histamine and PAF; this activation is actively regulated by Treg interactions. Macrophage activation by IgG also may represent an alternative pathway to PAF responses. Within the blood, neutrophil and basophil activation by either IgG or IgE presents additional pathways to generate these mediators if Ag becomes accessible. Kinetically slower, the release of preformed cytokines (TNF) or cytokines that are synthesized and then released (IL-33, IL-9) support localized tissue inflammation.

FIGURE 2.

Multiple pathways of anaphylaxis exist, mediated by either IgE or IgG and their respective FcRs. Within tissues, mast cell activation via IgE and FcεRI initiates early-phase responses mediated by histamine and PAF; this activation is actively regulated by Treg interactions. Macrophage activation by IgG also may represent an alternative pathway to PAF responses. Within the blood, neutrophil and basophil activation by either IgG or IgE presents additional pathways to generate these mediators if Ag becomes accessible. Kinetically slower, the release of preformed cytokines (TNF) or cytokines that are synthesized and then released (IL-33, IL-9) support localized tissue inflammation.

Close modal

In active sensitization models, IgE, FcεRI, and mast cells are responsible for inducing allergic diarrhea (58, 59). Although both allergen-specific IgE and IgG Abs are increased by sensitization, only FcεRI (58, 59), and not FcγRII/III (58), is required. Interestingly, the diarrhea response seems to be mediated by a combination of PAF and 5-HT. In contrast, the mast cell responses that are key in anaphylactic food allergy models (with contributions from macrophages and basophils) occur via IgE- and IgG-dependent mechanisms requiring both histamine and PAF (57, 61, 81). Recently, the necessity for basophils in peanut anaphylaxis also was defined (82). Interestingly, the pathways to systemic anaphylaxis models may relate to the Ag dose required to trigger each mechanism, because small doses activate the classical pathway, and large doses activate the alternative pathway (56).

Clinical studies showed that the incidence of food allergen–specific IgE is 10-fold greater than the incidence of food allergy (83), suggesting an additional level of tolerance regulation above that of simply preventing immunological priming toward Th2 and IgE. Furthermore, in patients with Stat3 mutations leading to hyper-IgE syndrome, anaphylactic reactivity to food allergens is actually diminished (84). Recent work showed that Tregs can suppress IgE-primed mast cell degranulation to Ag exposure via OX40/OX40 ligand interactions (85). In food allergy, we demonstrated that Treg transfer could suppress anaphylaxis and restore intestinal Th17 homeostasis by enhancing mast cell–derived IL-6 (41). Interestingly, this cytokine-mediated process was OX40 independent and instead was mediated via TGF-β (41). Additionally, Tregs can downregulate FcεRI on mast cells in vitro (86). This emerging form of active tolerance, occurring despite the presence of an Ag-specific IgE–primed immune system, seems distinct from Ag desensitization, which is associated with internalization of FcεRI and IgE and altered Syk activation (87, 88).

Immunologically, food allergy is a disease with much left to determine. The mechanisms of tolerance, both in terms of what prevents most people from developing responses, as well as why some individuals outgrow or never develop food allergies despite sensitization, remain unclear. Similarly, the environmental and genetic influences over sensitization are just becoming understood. Importantly, studies from animal models are showing that the mechanisms of anaphylactic reactions may well be heterogeneous in terms of routes of exposure, cell types involved, and the mediators responsible for symptoms. A better understanding of this heterogeneity will be crucial in developing future therapies.

This work was supported by funding from The Sunshine Foundation and National Institutes of Health Grant R01 AI076456.

Abbreviations used in this article:

DC

dendritic cell

5-HT

5-hydroxytryptamine

PAF

platelet-activating factor

SEB

staphylococcal enterotoxin B

Treg

regulatory T cell.

1
Gupta
R. S.
,
Springston
E. E.
,
Warrier
M. R.
,
Smith
B.
,
Kumar
R.
,
Pongracic
J.
,
Holl
J. L.
.
2011
.
The prevalence, severity, and distribution of childhood food allergy in the United States.
Pediatrics
128
:
e9
e17
.
2
Flokstra-de Blok
B. M.
,
Dubois
A. E.
,
Vlieg-Boerstra
B. J.
,
Oude Elberink
J. N.
,
Raat
H.
,
DunnGalvin
A.
,
Hourihane
J. O.
,
Duiverman
E. J.
.
2010
.
Health-related quality of life of food allergic patients: comparison with the general population and other diseases.
Allergy
65
:
238
244
.
3
Pons
L.
,
Ponnappan
U.
,
Hall
R. A.
,
Simpson
P.
,
Cockrell
G.
,
West
C. M.
,
Sampson
H. A.
,
Helm
R. M.
,
Burks
A. W.
.
2004
.
Soy immunotherapy for peanut-allergic mice: modulation of the peanut-allergic response.
J. Allergy Clin. Immunol.
114
:
915
921
.
4
Gharaibeh
T. M.
,
Safadi
R. A.
,
Rawashdeh
M. A.
,
Hammad
H. M.
.
2010
.
Plunging arteriovenous malformation in the floor of the mouth: a case report.
Br. J. Oral Maxillofac. Surg.
48
:
e35
e37
.
5
Burks
A. W.
,
Laubach
S.
,
Jones
S. M.
.
2008
.
Oral tolerance, food allergy, and immunotherapy: implications for future treatment.
J. Allergy Clin. Immunol.
121
:
1344
1350
.
6
Chehade
M.
,
Mayer
L.
.
2005
.
Oral tolerance and its relation to food hypersensitivities.
J. Allergy Clin. Immunol.
115
:
3
12, quiz 13
.
7
Pabst
O.
,
Mowat
A. M.
.
2012
.
Oral tolerance to food protein.
Mucosal Immunol.
5
:
232
239
.
8
Denning
T. L.
,
Wang
Y. C.
,
Patel
S. R.
,
Williams
I. R.
,
Pulendran
B.
.
2007
.
Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses.
Nat. Immunol.
8
:
1086
1094
.
9
Schulz
O.
,
Jaensson
E.
,
Persson
E. K.
,
Liu
X.
,
Worbs
T.
,
Agace
W. W.
,
Pabst
O.
.
2009
.
Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions.
J. Exp. Med.
206
:
3101
3114
.
10
Hadis
U.
,
Wahl
B.
,
Schulz
O.
,
Hardtke-Wolenski
M.
,
Schippers
A.
,
Wagner
N.
,
Müller
W.
,
Sparwasser
T.
,
Förster
R.
,
Pabst
O.
.
2011
.
Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria.
Immunity
34
:
237
246
.
11
Varol
C.
,
Vallon-Eberhard
A.
,
Elinav
E.
,
Aychek
T.
,
Shapira
Y.
,
Luche
H.
,
Fehling
H. J.
,
Hardt
W. D.
,
Shakhar
G.
,
Jung
S.
.
2009
.
Intestinal lamina propria dendritic cell subsets have different origin and functions.
Immunity
31
:
502
512
.
12
Jaensson
E.
,
Uronen-Hansson
H.
,
Pabst
O.
,
Eksteen
B.
,
Tian
J.
,
Coombes
J. L.
,
Berg
P. L.
,
Davidsson
T.
,
Powrie
F.
,
Johansson-Lindbom
B.
,
Agace
W. W.
.
2008
.
Small intestinal CD103+ dendritic cells display unique functional properties that are conserved between mice and humans.
J. Exp. Med.
205
:
2139
2149
.
13
Sun
C. M.
,
Hall
J. A.
,
Blank
R. B.
,
Bouladoux
N.
,
Oukka
M.
,
Mora
J. R.
,
Belkaid
Y.
.
2007
.
Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid.
J. Exp. Med.
204
:
1775
1785
.
14
Coombes
J. L.
,
Siddiqui
K. R.
,
Arancibia-Cárcamo
C. V.
,
Hall
J.
,
Sun
C. M.
,
Belkaid
Y.
,
Powrie
F.
.
2007
.
A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism.
J. Exp. Med.
204
:
1757
1764
.
15
Iwata
M.
,
Hirakiyama
A.
,
Eshima
Y.
,
Kagechika
H.
,
Kato
C.
,
Song
S. Y.
.
2004
.
Retinoic acid imprints gut-homing specificity on T cells.
Immunity
21
:
527
538
.
16
Cassani
B.
,
Villablanca
E. J.
,
Quintana
F. J.
,
Love
P. E.
,
Lacy-Hulbert
A.
,
Blaner
W. S.
,
Sparwasser
T.
,
Snapper
S. B.
,
Weiner
H. L.
,
Mora
J. R.
.
2011
.
Gut-tropic T cells that express integrin α4β7 and CCR9 are required for induction of oral immune tolerance in mice.
Gastroenterology
141
:
2109
2118
.
17
Mora
J. R.
,
Iwata
M.
,
Eksteen
B.
,
Song
S. Y.
,
Junt
T.
,
Senman
B.
,
Otipoby
K. L.
,
Yokota
A.
,
Takeuchi
H.
,
Ricciardi-Castagnoli
P.
, et al
.
2006
.
Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells.
Science
314
:
1157
1160
.
18
Matteoli
G.
,
Mazzini
E.
,
Iliev
I. D.
,
Mileti
E.
,
Fallarino
F.
,
Puccetti
P.
,
Chieppa
M.
,
Rescigno
M.
.
2010
.
Gut CD103+ dendritic cells express indoleamine 2,3-dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction.
Gut
59
:
595
604
.
19
Shan
M.
,
Gentile
M.
,
Yeiser
J. R.
,
Walland
A. C.
,
Bornstein
V. U.
,
Chen
K.
,
He
B.
,
Cassis
L.
,
Bigas
A.
,
Cols
M.
, et al
.
2013
.
Mucus enhances gut homeostasis and oral tolerance by delivering immunoregulatory signals.
Science
342
:
447
453
.
20
Bennett
C. L.
,
Christie
J.
,
Ramsdell
F.
,
Brunkow
M. E.
,
Ferguson
P. J.
,
Whitesell
L.
,
Kelly
T. E.
,
Saulsbury
F. T.
,
Chance
P. F.
,
Ochs
H. D.
.
2001
.
The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3.
Nat. Genet.
27
:
20
21
.
21
Torgerson
T. R.
,
Linane
A.
,
Moes
N.
,
Anover
S.
,
Mateo
V.
,
Rieux-Laucat
F.
,
Hermine
O.
,
Vijay
S.
,
Gambineri
E.
,
Cerf-Bensussan
N.
, et al
.
2007
.
Severe food allergy as a variant of IPEX syndrome caused by a deletion in a noncoding region of the FOXP3 gene.
Gastroenterology
132
:
1705
1717
.
22
Wildin
R. S.
,
Ramsdell
F.
,
Peake
J.
,
Faravelli
F.
,
Casanova
J. L.
,
Buist
N.
,
Levy-Lahad
E.
,
Mazzella
M.
,
Goulet
O.
,
Perroni
L.
, et al
.
2001
.
X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy.
Nat. Genet.
27
:
18
20
.
23
Kim
J. M.
,
Rasmussen
J. P.
,
Rudensky
A. Y.
.
2007
.
Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice.
Nat. Immunol.
8
:
191
197
.
24
Friedman
A.
,
Weiner
H. L.
.
1994
.
Induction of anergy or active suppression following oral tolerance is determined by antigen dosage.
Proc. Natl. Acad. Sci. USA
91
:
6688
6692
.
25
Ganeshan
K.
,
Neilsen
C. V.
,
Hadsaitong
A.
,
Schleimer
R. P.
,
Luo
X.
,
Bryce
P. J.
.
2009
.
Impairing oral tolerance promotes allergy and anaphylaxis: a new murine food allergy model.
J. Allergy Clin. Immunol.
123
:
231
-
238.e4
.
26
Shreffler
W. G.
,
Castro
R. R.
,
Kucuk
Z. Y.
,
Charlop-Powers
Z.
,
Grishina
G.
,
Yoo
S.
,
Burks
A. W.
,
Sampson
H. A.
.
2006
.
The major glycoprotein allergen from Arachis hypogaea, Ara h 1, is a ligand of dendritic cell-specific ICAM-grabbing nonintegrin and acts as a Th2 adjuvant in vitro.
J. Immunol.
177
:
3677
3685
.
27
Jyonouchi
S.
,
Abraham
V.
,
Orange
J. S.
,
Spergel
J. M.
,
Gober
L.
,
Dudek
E.
,
Saltzman
R.
,
Nichols
K. E.
,
Cianferoni
A.
.
2011
.
Invariant natural killer T cells from children with versus without food allergy exhibit differential responsiveness to milk-derived sphingomyelin.
J. Allergy Clin. Immunol.
128
:
102
-
109.e13
.
28
Berin
M. C.
,
Sampson
H. A.
.
2013
.
Mucosal immunology of food allergy.
Curr. Biol.
23
:
R389
400
.
29
Hazebrouck
S.
,
Przybylski-Nicaise
L.
,
Ah-Leung
S.
,
Adel-Patient
K.
,
Corthier
G.
,
Wal
J. M.
,
Rabot
S.
.
2009
.
Allergic sensitization to bovine beta-lactoglobulin: comparison between germ-free and conventional BALB/c mice.
Int. Arch. Allergy Immunol.
148
:
65
72
.
30
Hill
D. A.
,
Siracusa
M. C.
,
Abt
M. C.
,
Kim
B. S.
,
Kobuley
D.
,
Kubo
M.
,
Kambayashi
T.
,
Larosa
D. F.
,
Renner
E. D.
,
Orange
J. S.
, et al
.
2012
.
Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation.
Nat. Med.
18
:
538
546
.
31
Mathias
C. B.
,
Hobson
S. A.
,
Garcia-Lloret
M.
,
Lawson
G.
,
Poddighe
D.
,
Freyschmidt
E. J.
,
Xing
W.
,
Gurish
M. F.
,
Chatila
T. A.
,
Oettgen
H. C.
.
2011
.
IgE-mediated systemic anaphylaxis and impaired tolerance to food antigens in mice with enhanced IL-4 receptor signaling.
J. Allergy Clin. Immunol.
127
:
795
805.e1–6
.
32
Noval Rivas
M.
,
Burton
O. T.
,
Wise
P.
,
Zhang
Y. Q.
,
Hobson
S. A.
,
Garcia Lloret
M.
,
Chehoud
C.
,
Kuczynski
J.
,
DeSantis
T.
,
Warrington
J.
, et al
.
2013
.
A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis.
J. Allergy Clin. Immunol.
131
:
201
212
.
33
Hammad
H.
,
Lambrecht
B. N.
.
2008
.
Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma.
Nat. Rev. Immunol.
8
:
193
204
.
34
Chu
D. K.
,
Llop-Guevara
A.
,
Walker
T. D.
,
Flader
K.
,
Goncharova
S.
,
Boudreau
J. E.
,
Moore
C. L.
,
Seunghyun In
T.
,
Waserman
S.
,
Coyle
A. J.
, et al
.
2013
.
IL-33, but not thymic stromal lymphopoietin or IL-25, is central to mite and peanut allergic sensitization.
J. Allergy Clin. Immunol.
131
:
187
200.e1–8
.
35
Yang
Z.
,
Sun
R.
,
Grinchuk
V.
,
Fernández-Blanco
J. A.
,
Notari
L.
,
Bohl
J. A.
,
McLean
L. P.
,
Ramalingam
T. R.
,
Wynn
T. A.
,
Urban
J. F.
 Jr.
, et al
.
2013
.
IL-33-induced alterations in murine intestinal function and cytokine responses are MyD88, STAT6, and IL-13 dependent.
[Published erratum appears in 2013 Am. J. Physio. Gastrointest. Liver Physiol. 304: G1137.]
Am. J. Physiol. Gastrointest. Liver Physiol.
304
:
G381
G389
.
36
Turnquist
H. R.
,
Thomson
A. W.
.
2009
.
IL-33 broadens its repertoire to affect DC.
Eur. J. Immunol.
39
:
3292
3295
.
37
Tjota
M. Y.
,
Williams
J. W.
,
Lu
T.
,
Clay
B. S.
,
Byrd
T.
,
Hrusch
C. L.
,
Decker
D. C.
,
de Araujo
C. A.
,
Bryce
P. J.
,
Sperling
A. I.
.
2013
.
IL-33-dependent induction of allergic lung inflammation by FcγRIII signaling.
J. Clin. Invest.
123
:
2287
2297
.
38
Wills-Karp
M.
,
Rani
R.
,
Dienger
K.
,
Lewkowich
I.
,
Fox
J. G.
,
Perkins
C.
,
Lewis
L.
,
Finkelman
F. D.
,
Smith
D. E.
,
Bryce
P. J.
, et al
.
2012
.
Trefoil factor 2 rapidly induces interleukin 33 to promote type 2 immunity during allergic asthma and hookworm infection.
J. Exp. Med.
209
:
607
622
.
39
Berin
M. C.
,
Shreffler
W. G.
.
2008
.
T(H)2 adjuvants: implications for food allergy.
J. Allergy Clin. Immunol.
121
:
1311
-
1320, quiz 1321-1322
.
40
Yang
P. C.
,
Xing
Z.
,
Berin
C. M.
,
Soderholm
J. D.
,
Feng
B. S.
,
Wu
L.
,
Yeh
C.
.
2007
.
TIM-4 expressed by mucosal dendritic cells plays a critical role in food antigen-specific Th2 differentiation and intestinal allergy.
Gastroenterology
133
:
1522
1533
.
41
Ganeshan
K.
,
Bryce
P. J.
.
2012
.
Regulatory T cells enhance mast cell production of IL-6 via surface-bound TGF-β.
J. Immunol.
188
:
594
603
.
42
Berin
M. C.
,
Wang
W.
.
2013
.
Reduced severity of peanut-induced anaphylaxis in TLR9-deficient mice is associated with selective defects in humoral immunity.
Mucosal Immunol.
6
:
114
121
.
43
Groschwitz
K. R.
,
Hogan
S. P.
.
2009
.
Intestinal barrier function: molecular regulation and disease pathogenesis.
J. Allergy Clin. Immunol.
124
:
3
20, quiz 21–22
.
44
Yu
L. C. H.
,
Yang
P.-C.
,
Berin
M. C.
,
Di Leo
V.
,
Conrad
D. H.
,
McKay
D. M.
,
Satoskar
A. R.
,
Perdue
M. H.
.
2001
.
Enhanced transepithelial antigen transport in intestine of allergic mice is mediated by IgE/CD23 and regulated by interleukin-4.
Gastroenterology
121
:
370
381
.
45
Berin
M. C.
,
Mayer
L.
.
2009
.
Immunophysiology of experimental food allergy.
Mucosal Immunol.
2
:
24
32
.
46
Samuelov
L.
,
Sarig
O.
,
Harmon
R. M.
,
Rapaport
D.
,
Ishida-Yamamoto
A.
,
Isakov
O.
,
Koetsier
J. L.
,
Gat
A.
,
Goldberg
I.
,
Bergman
R.
, et al
.
2013
.
Desmoglein 1 deficiency results in severe dermatitis, multiple allergies and metabolic wasting.
Nat. Genet.
45
:
1244
1248
.
47
Sherrill
J. D.
,
Kc
K.
,
Wu
D.
,
Djukic
Z.
,
Caldwell
J. M.
,
Stucke
E. M.
,
Kemme
K. A.
,
Costello
M. S.
,
Mingler
M. K.
,
Blanchard
C.
, et al
.
2013
.
Desmoglein-1 regulates esophageal epithelial barrier function and immune responses in eosinophilic esophagitis.
Mucosal Immunol.
DOI: 10.1038/mi.2013.90.
48
Oyoshi
M. K.
,
Murphy
G. F.
,
Geha
R. S.
.
2009
.
Filaggrin-deficient mice exhibit TH17-dominated skin inflammation and permissiveness to epicutaneous sensitization with protein antigen.
J. Allergy Clin. Immunol.
124
:
485
-
493, 493.e1
.
49
Bartnikas
L. M.
,
Gurish
M. F.
,
Burton
O. T.
,
Leisten
S.
,
Janssen
E.
,
Oettgen
H. C.
,
Beaupre
J.
,
Lewis
C. N.
,
Austen
K. F.
,
Schulte
S.
, et al
.
2013
.
Epicutaneous sensitization results in IgE-dependent intestinal mast cell expansion and food-induced anaphylaxis.
J. Allergy Clin. Immunol.
131
:
451
460.e1–6
.
50
Brown
S. J.
,
Asai
Y.
,
Cordell
H. J.
,
Campbell
L. E.
,
Zhao
Y.
,
Liao
H.
,
Northstone
K.
,
Henderson
J.
,
Alizadehfar
R.
,
Ben-Shoshan
M.
, et al
.
2011
.
Loss-of-function variants in the filaggrin gene are a significant risk factor for peanut allergy.
J. Allergy Clin. Immunol.
127
:
661
667
.
51
Rothenberg
M. E.
,
Spergel
J. M.
,
Sherrill
J. D.
,
Annaiah
K.
,
Martin
L. J.
,
Cianferoni
A.
,
Gober
L.
,
Kim
C.
,
Glessner
J.
,
Frackelton
E.
, et al
.
2010
.
Common variants at 5q22 associate with pediatric eosinophilic esophagitis.
Nat. Genet.
42
:
289
291
.
52
Tan
T. H.
,
Ellis
J. A.
,
Saffery
R.
,
Allen
K. J.
.
2012
.
The role of genetics and environment in the rise of childhood food allergy.
Clin. Exp. Allergy
42
:
20
-
29
.
53
Ho
M. H.
,
Wong
W. H.
,
Chang
C.
.
2012
.
Clinical spectrum of food allergies: a comprehensive review.
Clin. Rev. Allergy Immunol.
DOI: 10.1007/s12016-012-8339-6.
54
Miyajima
I.
,
Dombrowicz
D.
,
Martin
T. R.
,
Ravetch
J. V.
,
Kinet
J. P.
,
Galli
S. J.
.
1997
.
Systemic anaphylaxis in the mouse can be mediated largely through IgG1 and Fc gammaRIII. Assessment of the cardiopulmonary changes, mast cell degranulation, and death associated with active or IgE- or IgG1-dependent passive anaphylaxis.
J. Clin. Invest.
99
:
901
914
.
55
Galli
S. J.
,
Tsai
M.
.
2012
.
IgE and mast cells in allergic disease.
Nat. Med.
18
:
693
704
.
56
Strait
R. T.
,
Morris
S. C.
,
Yang
M.
,
Qu
X. W.
,
Finkelman
F. D.
.
2002
.
Pathways of anaphylaxis in the mouse.
J. Allergy Clin. Immunol.
109
:
658
668
.
57
Sun
J.
,
Arias
K.
,
Alvarez
D.
,
Fattouh
R.
,
Walker
T.
,
Goncharova
S.
,
Kim
B.
,
Waserman
S.
,
Reed
J.
,
Coyle
A. J.
,
Jordana
M.
.
2007
.
Impact of CD40 ligand, B cells, and mast cells in peanut-induced anaphylactic responses.
J. Immunol.
179
:
6696
6703
.
58
Brandt
E. B.
,
Strait
R. T.
,
Hershko
D.
,
Wang
Q.
,
Muntel
E. E.
,
Scribner
T. A.
,
Zimmermann
N.
,
Finkelman
F. D.
,
Rothenberg
M. E.
.
2003
.
Mast cells are required for experimental oral allergen-induced diarrhea.
J. Clin. Invest.
112
:
1666
1677
.
59
Wang
M.
,
Takeda
K.
,
Shiraishi
Y.
,
Okamoto
M.
,
Dakhama
A.
,
Joetham
A.
,
Gelfand
E. W.
.
2010
.
Peanut-induced intestinal allergy is mediated through a mast cell-IgE-FcepsilonRI-IL-13 pathway.
J. Allergy Clin. Immunol.
126
:
306
316, 316.e1–12
.
60
Jönsson
F.
,
Mancardi
D. A.
,
Kita
Y.
,
Karasuyama
H.
,
Iannascoli
B.
,
Van Rooijen
N.
,
Shimizu
T.
,
Daëron
M.
,
Bruhns
P.
.
2011
.
Mouse and human neutrophils induce anaphylaxis.
J. Clin. Invest.
121
:
1484
1496
.
61
Arias
K.
,
Baig
M.
,
Colangelo
M.
,
Chu
D.
,
Walker
T.
,
Goncharova
S.
,
Coyle
A.
,
Vadas
P.
,
Waserman
S.
,
Jordana
M.
.
2009
.
Concurrent blockade of platelet-activating factor and histamine prevents life-threatening peanut-induced anaphylactic reactions.
J. Allergy Clin. Immunol.
124
:
307
314, 314.e1–2
.
62
Smit
J. J.
,
Willemsen
K.
,
Hassing
I.
,
Fiechter
D.
,
Storm
G.
,
van Bloois
L.
,
Leusen
J. H.
,
Pennings
M.
,
Zaiss
D.
,
Pieters
R. H.
.
2011
.
Contribution of classic and alternative effector pathways in peanut-induced anaphylactic responses.
PLoS ONE
6
:
e28917
.
63
Mancardi
D. A.
,
Albanesi
M.
,
Jönsson
F.
,
Iannascoli
B.
,
Van Rooijen
N.
,
Kang
X.
,
England
P.
,
Daëron
M.
,
Bruhns
P.
.
2013
.
The high-affinity human IgG receptor FcγRI (CD64) promotes IgG-mediated inflammation, anaphylaxis, and antitumor immunotherapy.
Blood
121
:
1563
1573
.
64
Vaz
N. M.
,
de Souza
C. M.
,
Hornbrook
M. M.
,
Hanson
D. G.
,
Lynch
N. R.
.
1977
.
Sensitivity to intravenous injections of histamine and serotonin in inbred mouse strains.
Int. Arch. Allergy Appl. Immunol.
53
:
545
554
.
65
Ishii
S.
,
Kuwaki
T.
,
Nagase
T.
,
Maki
K.
,
Tashiro
F.
,
Sunaga
S.
,
Cao
W. H.
,
Kume
K.
,
Fukuchi
Y.
,
Ikuta
K.
, et al
.
1998
.
Impaired anaphylactic responses with intact sensitivity to endotoxin in mice lacking a platelet-activating factor receptor.
J. Exp. Med.
187
:
1779
1788
.
66
Makabe-Kobayashi
Y.
,
Hori
Y.
,
Adachi
T.
,
Ishigaki-Suzuki
S.
,
Kikuchi
Y.
,
Kagaya
Y.
,
Shirato
K.
,
Nagy
A.
,
Ujike
A.
,
Takai
T.
, et al
.
2002
.
The control effect of histamine on body temperature and respiratory function in IgE-dependent systemic anaphylaxis.
J. Allergy Clin. Immunol.
110
:
298
303
.
67
Wechsler
J. B.
,
Schroeder
H. A.
,
Byrne
A. J.
,
Chien
K. B.
,
Bryce
P. J.
.
2013
.
Anaphylactic responses to histamine in mice utilize both histamine receptors 1 and 2.
Allergy
68
:
1338
1340
.
68
Lin
R. Y.
,
Curry
A.
,
Pesola
G. R.
,
Knight
R. J.
,
Lee
H. S.
,
Bakalchuk
L.
,
Tenenbaum
C.
,
Westfal
R. E.
.
2000
.
Improved outcomes in patients with acute allergic syndromes who are treated with combined H1 and H2 antagonists.
Ann. Emerg. Med.
36
:
462
468
.
69
McManus
L. M.
,
Pinckard
R. N.
.
2000
.
PAF, a putative mediator of oral inflammation.
Crit. Rev. Oral Biol. Med.
11
:
240
258
.
70
Ogawa
Y.
,
Grant
J. A.
.
2007
.
Mediators of anaphylaxis.
Immunol. Allergy Clin. North Am.
27
:
249
260, vii
.
71
Wershil
B. K.
,
Wang
Z. S.
,
Gordon
J. R.
,
Galli
S. J.
.
1991
.
Recruitment of neutrophils during IgE-dependent cutaneous late phase reactions in the mouse is mast cell-dependent. Partial inhibition of the reaction with antiserum against tumor necrosis factor-alpha.
J. Clin. Invest.
87
:
446
453
.
72
Hsu
C. L.
,
Neilsen
C. V.
,
Bryce
P. J.
.
2010
.
IL-33 is produced by mast cells and regulates IgE-dependent inflammation.
PLoS ONE
5
:
e11944
.
73
Choi
I. W.
,
Kim
Y. S.
,
Kim
D. K.
,
Choi
J. H.
,
Seo
K. H.
,
Im
S. Y.
,
Kwon
K. S.
,
Lee
M. S.
,
Ha
T. Y.
,
Lee
H. K.
.
2003
.
Platelet-activating factor-mediated NF-kappaB dependency of a late anaphylactic reaction.
J. Exp. Med.
198
:
145
151
.
74
Jung
M. Y.
,
Smrž
D.
,
Desai
A.
,
Bandara
G.
,
Ito
T.
,
Iwaki
S.
,
Kang
J. H.
,
Andrade
M. V.
,
Hilderbrand
S. C.
,
Brown
J. M.
, et al
.
2013
.
IL-33 induces a hyporesponsive phenotype in human and mouse mast cells.
J. Immunol.
190
:
531
538
.
75
Bouffi
C.
,
Rochman
M.
,
Zust
C. B.
,
Stucke
E. M.
,
Kartashov
A.
,
Fulkerson
P. C.
,
Barski
A.
,
Rothenberg
M. E.
.
2013
.
IL-33 markedly activates murine eosinophils by an NF-κB-dependent mechanism differentially dependent upon an IL-4-driven autoinflammatory loop.
J. Immunol.
191
:
4317
4325
.
76
Goswami
R.
,
Kaplan
M. H.
.
2011
.
A brief history of IL-9.
J. Immunol.
186
:
3283
3288
.
77
Forbes
E. E.
,
Groschwitz
K.
,
Abonia
J. P.
,
Brandt
E. B.
,
Cohen
E.
,
Blanchard
C.
,
Ahrens
R.
,
Seidu
L.
,
McKenzie
A.
,
Strait
R.
, et al
.
2008
.
IL-9- and mast cell-mediated intestinal permeability predisposes to oral antigen hypersensitivity.
J. Exp. Med.
205
:
897
913
.
78
Osterfeld
H.
,
Ahrens
R.
,
Strait
R.
,
Finkelman
F. D.
,
Renauld
J. C.
,
Hogan
S. P.
.
2010
.
Differential roles for the IL-9/IL-9 receptor alpha-chain pathway in systemic and oral antigen-induced anaphylaxis.
J. Allergy Clin. Immunol.
125
:
469
476.e2
.
79
Finkelman
F. D.
2007
.
Anaphylaxis: lessons from mouse models.
J. Allergy Clin. Immunol.
120
:
506
515; quiz 516–7
.
80
Tsujimura
Y.
,
Obata
K.
,
Mukai
K.
,
Shindou
H.
,
Yoshida
M.
,
Nishikado
H.
,
Kawano
Y.
,
Minegishi
Y.
,
Shimizu
T.
,
Karasuyama
H.
.
2008
.
Basophils play a pivotal role in immunoglobulin-G-mediated but not immunoglobulin-E-mediated systemic anaphylaxis.
Immunity
28
:
581
589
.
81
Arias
K.
,
Chu
D. K.
,
Flader
K.
,
Botelho
F.
,
Walker
T.
,
Arias
N.
,
Humbles
A. A.
,
Coyle
A. J.
,
Oettgen
H. C.
,
Chang
H. D.
, et al
.
2011
.
Distinct immune effector pathways contribute to the full expression of peanut-induced anaphylactic reactions in mice.
J. Allergy Clin. Immunol.
127
:
1552
1561.e1
.
82
Reber
L. L.
,
Marichal
T.
,
Mukai
K.
,
Kita
Y.
,
Tokuoka
S. M.
,
Roers
A.
,
Hartmann
K.
,
Karasuyama
H.
,
Nadeau
K. C.
,
Tsai
M.
,
Galli
S. J.
.
2013
.
Selective ablation of mast cells or basophils reduces peanut-induced anaphylaxis in mice.
J. Allergy Clin. Immunol.
132
:
881
888.e1–11
.
83
Liu
A. H.
,
Jaramillo
R.
,
Sicherer
S. H.
,
Wood
R. A.
,
Bock
S. A.
,
Burks
A. W.
,
Massing
M.
,
Cohn
R. D.
,
Zeldin
D. C.
.
2010
.
National prevalence and risk factors for food allergy and relationship to asthma: results from the National Health and Nutrition Examination Survey 2005-2006.
J. Allergy Clin. Immunol.
126
:
798
806.e13
.
84
Siegel
A. M.
,
Stone
K. D.
,
Cruse
G.
,
Lawrence
M. G.
,
Olivera
A.
,
Jung
M. Y.
,
Barber
J. S.
,
Freeman
A. F.
,
Holland
S. M.
,
O'Brien
M.
, et al
.
2013
.
Diminished allergic disease in patients with STAT3 mutations reveals a role for STAT3 signaling in mast cell degranulation.
J. Allergy Clin. Immunol.
132
:
1388
1396
.
85
Gri
G.
,
Piconese
S.
,
Frossi
B.
,
Manfroi
V.
,
Merluzzi
S.
,
Tripodo
C.
,
Viola
A.
,
Odom
S.
,
Rivera
J.
,
Colombo
M. P.
,
Pucillo
C. E.
.
2008
.
CD4+CD25+ regulatory T cells suppress mast cell degranulation and allergic responses through OX40-OX40L interaction.
Immunity
29
:
771
781
.
86
Kashyap
M.
,
Thornton
A. M.
,
Norton
S. K.
,
Barnstein
B.
,
Macey
M.
,
Brenzovich
J.
,
Shevach
E.
,
Leonard
W. J.
,
Ryan
J. J.
.
2008
.
Cutting edge: CD4 T cell-mast cell interactions alter IgE receptor expression and signaling.
J. Immunol.
180
:
2039
2043
.
87
Oka
T.
,
Rios
E. J.
,
Tsai
M.
,
Kalesnikoff
J.
,
Galli
S. J.
.
2013
.
Rapid desensitization induces internalization of antigen-specific IgE on mouse mast cells.
J. Allergy Clin. Immunol.
132
:
922
932.e1–16
.
88
Khodoun
M. V.
,
Kucuk
Z. Y.
,
Strait
R. T.
,
Krishnamurthy
D.
,
Janek
K.
,
Lewkowich
I.
,
Morris
S. C.
,
Finkelman
F. D.
.
2013
.
Rapid polyclonal desensitization with antibodies to IgE and FcεRIα.
J. Allergy Clin. Immunol.
131
:
1555
1564
.

The authors have no financial conflicts of interest.