ArticlePDF Available

The Effect of Digestion and Digestibility on Allergenicity of Food

MDPI
Nutrients
Authors:

Abstract and Figures

Food allergy prevalence numbers are still on the rise. Apart from environmental influences, dietary habits, food availability and life-style factors, medication could also play a role. For immune tolerance of food, several contributing factors ensure that dietary compounds are immunologically ignored and serve only as source for energy and nutrient supply. Functional digestion along the gastrointestinal tract is essential for the molecular breakdown and a prerequisite for appropriate uptake in the intestine. Digestion and digestibility of carbohydrates and proteins thus critically affect the risk of food allergy development. In this review, we highlight the influence of amylases, gastric acid- and trypsin-inhibitors, as well as of food processing in the context of food allergenicity.
This content is subject to copyright.
nutrients
Review
The Effect of Digestion and Digestibility on
Allergenicity of Food
Isabella Pali-Schöll 1, 2, *, Eva Untersmayr 2, Martina Klems 2and Erika Jensen-Jarolim 1,2
1Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary
Medicine Vienna, Medical University Vienna and University Vienna, Veterinärplatz 1, 1210 Vienna, Austria;
erika.jensen-jarolim@meduniwien.ac.at
2Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and
Immunology, Medical University of Vienna, Währinger Gürtel 18–20, 1090 Vienna, Austria;
eva.untersmayr@meduniwien.ac.at (E.U.); martina.klems@meduniwien.ac.at (M.K.)
*Correspondence: isabella.pali@vetmeduni.ac.at; Tel.: +43-664-60257-6259
Received: 29 June 2018; Accepted: 13 August 2018; Published: 21 August 2018


Abstract:
Food allergy prevalence numbers are still on the rise. Apart from environmental influences,
dietary habits, food availability and life-style factors, medication could also play a role. For immune
tolerance of food, several contributing factors ensure that dietary compounds are immunologically
ignored and serve only as source for energy and nutrient supply. Functional digestion along the
gastrointestinal tract is essential for the molecular breakdown and a prerequisite for appropriate
uptake in the intestine. Digestion and digestibility of carbohydrates and proteins thus critically affect
the risk of food allergy development. In this review, we highlight the influence of amylases, gastric
acid- and trypsin-inhibitors, as well as of food processing in the context of food allergenicity.
Keywords:
anti-acid; acid suppressing medication; bariatric surgery; blocked digestion; food allergy;
gastritis; impaired digestion; Maillard; reflux; ulcer
1. Introduction
The prevalence of adverse reactions to food is still increasing. In the United States, an estimated
rise from about 3% (1997–1999) to 6% (2016) of children younger than 18 years affected by food allergies
has been reported [
1
]. A number of different factors are discussed to influence the development of
food allergies. Among these factors are smoking incl. passive or second-hand smoke [
2
], the changed
environment and/or pollution [
3
], altered vitamin D levels [
4
], and dual allergen exposure (skin
contact with food proteins compared to oral exposure) [
5
]. Also, increased hygiene resulting in
reduced microbiota diversity [
6
] or usage of antibiotics early in life disturbing the microbial balance in
the intestine [
7
] seem to play a role. Furthermore, the diet of the mother during pregnancy/lactation [
8
],
and additionally, the type and time point of complementary food introduction for the child could
be important [
9
]. For the latter, the recommendations have been updated recently [
10
], now stating
that introduction of allergenic food, e.g., peanuts, even to high-risk children should not be avoided or
postponed [1113].
Furthermore, digestion and digestibility could determine whether food proteins are tolerated or
become sensitizing agents. This aspect has therefore even been taken up by the European Food Safety
Agency in their scientific opinion about evaluation of allergenicity of food and feed proteins. Higher
resistance to digestion or survival along the digestive tract seems to increase the sensitization capacity
of a food component and renders it more immunogenic and/or allergenic. Based on this scientific
background, the present review article highlights factors influencing protein digestion and digestibility.
Nutrients 2018,10, 1129; doi:10.3390/nu10091129 www.mdpi.com/journal/nutrients
Nutrients 2018,10, 1129 2 of 16
2. Digestion of Carbohydrates: Amylase Action Critical for Starch Digestion and Microbiome
In green plants, starch accumulates as a product of photosynthesis. As a complex polysaccharide,
it represents a significant compound of our diet and serves as energy supply, but also as food
matrix. Also the food industry takes advantage of starch by supplementing it to infant food for
maintaining “colonic health” [
14
]. Starch is digested by specific enzymes, i.e., amylases, which cleave
the
α
-1,4-glucosidic bond of its major compound amylose, as well as the
α
-1,6-glucosidic bond of
the second major constituent, amylopectin [
15
]. In microbes, the amylase enzyme group consists
of 19 members, each with unique catalytic properties. They are technically applied in the starch
saccharification industry [
15
]. However, transient malabsorption due to immaturity of the GIT during
growth of the young child must be taken into consideration [16].
It is important to understand the biological impact of amylases, which are well conserved in
the animal kingdom [
17
]. In humans,
α
-amylase is a product of the exocrine pancreas. Animal
models suggest that microbial amylases could be supplied in pancreas insufficiency [
18
]. It is not
known whether this will be linked to a risk for sensitization, but
α
-amylase per se when inhaled is a
well-known occupational allergen. In baker’s asthma associated with the flour processing industry,
allergenic amylase derives from contaminating fungi [19].
In mammals, amylase is also secreted into the saliva. Its role in starch digestion has been
questioned due to its low amount relative to the overall amylase activity [
20
]. However,
in vitro
studies strongly propose that salivary amylolytic activity hydrolyzes up to 80% of bread starch in the
first 30 min of gastric digestion, independent of acidification by the gastric juices [
21
]. This critically
affects the quality of remnants reaching the intestine, which will affect the composition of the microflora
(discussed below).
While in human medicine this is less known, psychologists take advantage of salivary amylase
as a non-invasive biomarker for the evaluation of acute stress response [
22
] and it is increasingly
used in behavioral medicine [
23
]. The biological relevance of this phenomenon might be a need
of quick energy supply in form of glucose in the “fight-or-flight” reaction. Biomarker research
indicates that stress also has an effect on immune reactions. For instance, the release of salivary
α
-amylase indicated that experimental stress was higher in rural participants raised in the presence of
animals [
24
]. Acute or chronic stress may therefore quantitatively regulate amylase activity, and thereby
impact on the composition of digested carbohydrates and subsequently affect microbiota composition
(discussed below).
The amylase action on rapidly digestible starch (RDS) renders smaller products, like disaccharides
and trisaccharides [
25
]. These are then further hydrolyzed to glucose by other enzymes, such as
α
-glucosidase in the small intestine [
26
]. However, both amylase and
α
-glucosidase may act
synergistically. Some compounds represent slow-digestible starch (SDS), or resistant starch (RS)
as larger leftovers, which persist the gastrointestinal transit to a large degree. Usually, resulting levels
of malto-oligosaccharide indicate the degree of granular starch breakdown.
The starch breakdown by amylases is largely influenced by the composition of the food processing
and matrix composition. Cooking has been shown to enhance the amylase breakdown of starch [
27
],
which also depended on the individual
α
-amylase activity. Flavonoids are important plant constituents,
which interfere with amylase activity by hydrophobic interaction in the food matrix or by formation of
covalent bonds during cooking or in gastric juice, and therefore impair starch digestion [
28
]. This opens
up potential intervention strategies in diabetic patients to decrease the fermentation speed of starch
and thereby inhibit an undesired fast release of glucose. Starch may also form complexes with
lipids in the food matrix, e.g., complex formation with palm oil interfered with the digestion of rice
starches [
29
]. Interestingly, some fresh food may neutralize amylases by proteolysis. Kiwi contains
actinidin, a cysteine proteinase, which specifically attacks amylase and thereby may inhibit starch
digestion [30]. This may affect the presentation of allergenic epitopes in the food matrix.
Amylase in the duodenum also plays a key role in the breakdown of gluten and may therefore
modulate its pathophysiologic role in celiac disease [
31
]. While starch forms complexes with gluten
Nutrients 2018,10, 1129 3 of 16
during baking of bread, amylase resolves them and makes gluten accessible for thorough protein
digestion. Wheat on the other hand contains anti-enzymes, such as the ATIs (amylase-trypsin
inhibitors) with a role in non-celiac gluten sensitivity (NCGS) [
32
]. Nutritional ATIs additionally
stimulate the innate immune reaction via TLR4 [
32
] and thereby exacerbate allergic inflammation
not only in the intestine, but also in the airways in mouse models [
33
,
34
]. It is hypothesized that
industrial food processing contributes to the increased numbers of non-celiac gluten/wheat sensitivity
by stabilizing e.g., starch-gluten complexes, thereby bypassing the salivary and pancreatic enzymes,
leaving the digestion to mucosal amylases [35].
Processing may also affect the nanostructure of food, again affecting the amylase fermentation
and hydrolyzed products thereof. Depending on the composition, the RS fraction can serve as a
form of “prebiotics” fostering a bacterial community with benefits for health [
36
,
37
], confirmed
recently in an animal model [
38
]. Dietary inclusion of RS changed the 16S rRNA profiles of the gene
bacterial community, the profile of short-chain fatty acids (SCFA) and the overall lipid metabolism in
pigs [
39
]. In humans, a high RS proportion resulted in a beneficial increase in the ratio of Firmicutes to
Bacteroidetes [
40
], in favor of immune protection against allergies [
41
]. Therefore, starch digestion via
modulating microbiota richness also impacts food allergy.
Overall, starch is a major nutrient compound and food matrix, and industrial processing critically
interferes with its fermentation by amylase. Physiologically, stress enhances salivary amylase release,
and pancreatic disorders are associated with polysaccharide maldigestion. Both, starch and amylase
activity have implication for energy supply and the composition of RS remnants, which again critically
affect microbiota composition.
Extracting the evidence from all aspects of pathophysiological starch digestion in correlation with
life-style factors, we anticipate that amylase action may have an impact on the allergenicity of food by
several means:
(1)
It may result in epitope modification of plant food allergens or reveal neo-epitopes;
(2)
Starch and other food matrix compounds may form stable complexes during food processing,
supporting the transit of intact allergens;
(3)
Amylase action affects the composition of fermentation products with significant effect on
microbiota composition.
Presumably, this has impact not only on the control of celiac and non-celiac gluten
hypersensitivity [
32
], but also on type I food allergy with early life being critical [
42
]. More studies
need to be done to understand how exactly the microbiome could be manipulated in allergy and
asthma [43], but targeting starch digestion could be an interesting option.
3. Digestion and Digestibility of Proteins Associated with Lipids or Carbohydrates
There is general agreement that resistance of proteins to gastric digestion is an indicator for
potential allergenicity. For instance, in an vivo rat model digested vs. non-digested BLG was compared,
and clearly the intact BLG induced more IgE, IgA, and IgG1, linking the digestion and digestibility of
BLG directly to allergenicity [
44
]. This implies that any condition that keeps a certain protein intact
adds to the risk of food allergy induction.
A very important family among allergens are lipid-transfer proteins (LTPs). It was recently shown
that ligand binding can have different effects on their
in vitro
digestibility. In most cases, binding of
lipids to LTPs increases resistance to digestion. This was for instance shown for LTP from peach and
sunflower [
45
]. Sunflower seed was reported to be the most frequent elicitor of severe allergic reactions
in Europe, even more frequent than peanut, and listed in the middle field of food sensitizations in
European adults [
46
]. The LTP is stabilized against gastric digestion when phosphatidylcholine (PC)
was added
in vitro
. However,
in vivo
proof that PC-stabilization also leads to increased allergenicity
of the LTP so far is missing. Furthermore, the influence of (impaired) gastric vs. duodenal/intestinal
digestion needs to be investigated for oral sensitization in animal models.
Nutrients 2018,10, 1129 4 of 16
Furthermore, binding of lipids like PC to
β
-lactoglobulin and
α
-lactalbumin interferes with their
digestibility [
47
,
48
]. However, sensitization studies, which directly compare allergen with or without
attached lipid in vivo, are lacking.
Peptic digestion of grape LTP was not influenced by the presence of PC, and both molecules (with
or without PC) were able to induce skin prick test reactivity in allergic patients. It seems important
that the grape LTP is very stable to pepsin digestion, with or without the presence of lipid.
Similarly, binding of linoleic acid to wheat LTP did not change the gastric digestibility, and only
slightly increased its susceptibility to gastroduodenal digestion via changes in the structure [
49
]. Wheat
LTP was described as an important protein recognized by patients with food allergy to wheat [
50
],
and in around 60% of people with baker’s asthma, although it elicits sensitization via the respiratory
tract, where digestion might not play a crucial role [51].
In addition to individual molecules such as lipids the overall food matrix may play a crucial
role in the availability of different proteins for enzymatic breakdown, as was shown e.g., for peanut
allergens [52].
Apart from loading with different molecules and additional effects of food matrix, food processing
may change the digestibility and allergenicity of food [
53
]. Pasteurization of milk is very common
and important. However, this heating process can cause aggregation of food proteins such as
β
-lactoglobulin and
α
-lactalbumin. This aggregation was shown to enhance the uptake by Peyer’s
patches and Th2-mediated antibody and cytokine production in mice [54].
During the heating of food products, which contain sugars and proteins, not only does aggregation
of proteins occur, but so does the so-called Maillard reaction (MR). This reaction leads to products
that are responsible for color, flavor and taste e.g., in many fast food products, bakery products or
roasted peanuts. During this non-enzymatic reaction, free amino groups (mainly lysine and arginine)
of protein side chains can be occupied by covalent binding of reducing sugars, i.e., glycation. Schiff
bases are formed, followed by Amadori rearrangement and oxidative processes, altogether responsible
for the formation of advanced glycation end products (AGE), a chemically heterogeneous and unstable
group of molecules. These processes lead to a modified availability of enzymatic cleavage sites of
the protein. Higher glycation of
β
-lactoglobulin resulted in reduced susceptibility to digestion by
trypsin/chymotrypsin [
55
]. Glycation also decreased
in vitro
digestibility for patatin from potato [
56
],
tropomyosin from scallop [
57
], and high-molecular weight peanut proteins such as Ara h 1 [
58
].
The peanut proteins were more resistant to digestion in the fried and roasted peanuts than in the raw
and boiled samples. Also, wheat flour proteins in bread crumb and crust gained higher resistance
and IgE-binding capacity during the baking process [
59
], as did the glycated egg protein ovalbumin,
but not ovomucoid [
60
]. The latter study also showed a time-dependency during formation of MP,
as OVA glycated for 96 h was much more stable to gastric and duodenal digestion than OVA glycated
for 48 h.
Contrasting to these results with OVA, walnut 11S globulin after heating/roasting [
61
],
and lysozyme and codfish parvalbumin after glycation [
62
,
63
] ended up in higher solubility and
digestibility. These divergent observations may be explained by the different internal structural
characteristics of the proteins, and additionally by different types of sugar used in the MR:
galactose-glycation of
β
-lactoglobulin resulted in higher digestion-resistance than glycation with
tagatose [64].
The AGE per se can also be found in plasma and urine correlating to the amount in the
diet [
65
]. They can also be used by human microbiota of the lower gastrointestinal tract as energy
source [
66
], and probably modify the microbiota composition. The appearance in the GIT and
blood system makes AGE also likely to interact with immune cells, for instance activation of DC
via AGE-receptors (AGE-receptor complex, scavenger receptors A and B, mannose receptor, CD36;
reviewed in Ref. [
67
]) was shown [
68
]. Binding of roasted Ara h 1 was shown to occur to scavenger
receptor CD36 and receptor for advanced glycated end products RAGE [
69
,
70
], and for roasted Ara h
3 to mannose receptor [
71
]. Most importantly, this engagement leads to cellular signaling resulting
Nutrients 2018,10, 1129 5 of 16
in pro-inflammatory responses and enhanced allergic sensitization, as was shown in a mouse model
comparing raw vs. roasted peanut [
69
]. The animals sensitized with dry-roasted peanut extract
showed higher IL-4, IL-5, and IL-13 levels, as well as more specific IgG- and IgE-antibodies and
degranulation of effector cells. The usage of AGE-modified OVA (compared to native OVA) proved
that the NF
κ
B-pathway of DCs is involved in this outcome, as well as more efficient activation of
OVA-specific CD4+ T-cells, releasing more Th2-specific cytokines like IL-4, IL-5 and IL-6 [
72
,
73
].
Overall, AGEs and more specifically glycated food allergens may have enhanced T-cell activation
potential and thereby could increase the risk for allergic sensitization and/or effector cell reactions
(reviewed in [74]).
In vivo
data gathered with Maillard products and the effect on allergenicity are scarce. In humans,
a diet rich in MR-products (MRP), like AGE, limited the digestion of the protein. This was shown
in healthy young males as appearance of higher fecal nitrogen, lower absorbed nitrogen, and lower
digestibility of nitrogen [
75
]. Animal models show different
in vivo
effects of MRP-application
regarding the allergenicity. Depending on the conditions used, the capacity of the protein to evoke
sensitization and/or allergic reactions in Balb/c mice increased for AGE-OVA [
76
] and for roasted
peanuts [
69
]. In contrast, there was a decrease in sensitization potential for glycated tropomyosin
and arginine kinase from crustaceans [
77
], for buckwheat allergen Fag e 1 [
78
], and for chickpea
protein [79].
Finally, Maillard products also display an altered recognition by specific IgE present in allergic
patients or animals. This might be due to (i) the changes in the tertiary and secondary structure,
which can disrupt conformational or linear epitopes and reduce IgE-binding [
80
], (ii) formation of
aggregates, which show enhanced degranulation [
81
], and (iii) formation of new IgE-epitopes, as was
shown for Pecan nut, wheat flour and soybean. These foods only induced allergic reactions after
cooking, long storage or heating [
82
,
83
]. Whereas important allergens from cherry (Pru av 1) [
84
],
hazelnut (Cor a 1) [
85
], and milk (
β
-lactoglobulin) [
86
] showed reduced IgE-binding after heating in
presence of (poly)saccharides, the allergens from peanut (Ara h 1 and Ara h 2) displayed significantly
higher IgE-binding after non-enzymatic browning [87].
Importantly, it is necessary to define the final allergenicity of roasted food
in vivo
, as among 17
hazelnut-allergic patients, 5 still had positive DBPCFC-reactions, even though other methods (SPT,
HR, specific IgE-binding) showed a reduced allergenicity of the roasted form of hazelnut [88].
Taken together, the heating of foods which contain reducing sugars together with proteins leads to
the Maillard reaction and changes the conformation of the protein. This process can lead to (i) different
digestibility of some proteins, (ii) masking of existing antibody epitopes, or (iii) formation of novel
molecules, and may thereby also modify the immunogenicity and allergenicity of food proteins
(reviewed in [
74
,
89
]). The resulting immunoreactivity of glycated proteins may decrease, remain
unchanged, or even increase after food glycation [67].
With certainty, further studies are warranted to show the effects of the Maillard reaction for
individual, structural diverse protein molecules, different sugars, the dependency on temperature, pH,
duration of processing, water content and activity of the product, and probably also the food matrix.
The effect on digestibility and subsequent immunogenicity and allergenicity has to be shown
in vivo
.
4. Digestion of Proteins: Gastric Acid is Critical for Adequate Protein Digestion and Prevention
of Food Allergy
The digestibility of antigens has since long been considered a critical prerequisite for the induction
of food allergy [
90
]. However, also a number of digestion-labile proteins were shown to induce allergic
symptoms by primary sensitization without any co-existing pollen allergy, for instance hazelnut [91].
Digestion of proteins -and therefore most food allergens- is initiated in the stomach. A low
pH is essential for the inactive enzyme pepsinogen to get activated into pepsin [
92
]. However,
if acid-suppressing drugs are given, the pH increases considerably (e.g., up to 5 with proton pump
inhibitors, PPI).
Nutrients 2018,10, 1129 6 of 16
As shown in many previous
in vitro
experiments, the proper digestion by pepsin is hindered
when the pH is increased (Figure 1), and this is true for a number of food proteins, like hazelnut [
93
],
codfish [94], milk [95], and casein (Figure 1).
Nutrients 2018, 10, x FOR PEER REVIEW 6 of 15
As shown in many previous in vitro experiments, the proper digestion by pepsin is hindered
when the pH is increased (Figure 1), and this is true for a number of food proteins, like hazelnut [93],
codfish [94], milk [95], and casein (Figure 1).
Figure 1. (A) Digestion of proteins is hampered when pH increases. Proteins, as part of the daily diet,
are digested at low pH and broken down into smaller fragments, whereas a higher pH blocks proper
digestion. The resulting bigger fragments or proteins are more easily recognized by the immune
system, leading to an increased risk for sensitization or allergic reactions. (B) Digestion of α-casein in
vitro is hampered when pH increases. Casein was readily broken down by enzymatic digestion with
pepsin at pH 2.0, but remained totally intact even after 2 h of incubation with enzyme at pH 5.0. M:
molecular weight marker; -: empty lane; P: pepsin; 0: no incubation time, reaction stopped
immediately;: seconds; ‘: minutes; h: hour(s); Cas: casein.
It is clear that food intake per se changes the gastric pH, which can increase from a median
fasting baseline value of pH 1 to pH 4.5 with ingestion of the meal [96]. The buffer capacity thereby
depends on the food composition and meal constituents. However, this effect is transient, as
ongoing acid production is responsible for a subsequent decrease of the pH, which returns to ca. pH
1 about 260 min after the start of the meal [96]. Applying acid-suppressing substances can disturb
this process and induce a long-lasting elevation of the gastric pH up to 5.0 [97].
In a number of food animal models, the effect of this pH-elevation was shown in vivo, as
feeding digestion-labile antigen under concomitant acid-suppression resulted in a clear
Th2-response and allergy symptoms [98–104].
This acquired sensitization capacity was true for different proteins, like codfish, hazelnut or
ovalbumin, and even oral drugs, in the mouse model [99] and also in humans [105]. Importantly,
several types of acid-suppressing or -neutralizing medication, like base powder [106], sucralfate
[102], H2-receptor blockers [107] and proton pump inhibitors [101] produced this effect. The
outcome of the immune response may depend on timing of the anti-acid drug application in relation
to food uptake, and on the dosage of the antigen [101,108].
Gastric acid suppression might further impact on intestinal pH levels and consequently on
protein digestion in the intestine [109]. This assumption, however, requires further investigations in
clinical settings.
Undoubtedly, knowledge derived from experimental as well as in vitro studies simulating
human gastric digestion has to be confirmed using human samples and should be preferentially
translated into a clinical setting to confirm the relevance for patients. In 1992, Burks and coauthors
reported a 100-fold and 10-fold reduced IgE binding capacity of peanut and soybean allergens,
Figure 1.
(
A
) Digestion of proteins is hampered when pH increases. Proteins, as part of the daily
diet, are digested at low pH and broken down into smaller fragments, whereas a higher pH blocks
proper digestion. The resulting bigger fragments or proteins are more easily recognized by the immune
system, leading to an increased risk for sensitization or allergic reactions. (
B
) Digestion of
α
-casein
in vitro
is hampered when pH increases. Casein was readily broken down by enzymatic digestion
with pepsin at pH 2.0, but remained totally intact even after 2 h of incubation with enzyme at pH
5.0. M: molecular weight marker; -: empty lane; P: pepsin; 0: no incubation time, reaction stopped
immediately; “: seconds; ‘: minutes; h: hour(s); Cas: casein.
It is clear that food intake per se changes the gastric pH, which can increase from a median fasting
baseline value of pH 1 to pH 4.5 with ingestion of the meal [
96
]. The buffer capacity thereby depends
on the food composition and meal constituents. However, this effect is transient, as ongoing acid
production is responsible for a subsequent decrease of the pH, which returns to ca. pH 1 about 260 min
after the start of the meal [
96
]. Applying acid-suppressing substances can disturb this process and
induce a long-lasting elevation of the gastric pH up to 5.0 [97].
In a number of food animal models, the effect of this pH-elevation was shown
in vivo
, as feeding
digestion-labile antigen under concomitant acid-suppression resulted in a clear Th2-response and
allergy symptoms [98104].
This acquired sensitization capacity was true for different proteins, like codfish, hazelnut or
ovalbumin, and even oral drugs, in the mouse model [
99
] and also in humans [
105
]. Importantly,
several types of acid-suppressing or -neutralizing medication, like base powder [
106
], sucralfate [
102
],
H2-receptor blockers [
107
] and proton pump inhibitors [
101
] produced this effect. The outcome of the
immune response may depend on timing of the anti-acid drug application in relation to food uptake,
and on the dosage of the antigen [101,108].
Gastric acid suppression might further impact on intestinal pH levels and consequently on
protein digestion in the intestine [
109
]. This assumption, however, requires further investigations in
clinical settings.
Undoubtedly, knowledge derived from experimental as well as
in vitro
studies simulating human
gastric digestion has to be confirmed using human samples and should be preferentially translated into
Nutrients 2018,10, 1129 7 of 16
a clinical setting to confirm the relevance for patients. In 1992, Burks and coauthors reported a 100-fold
and 10-fold reduced IgE binding capacity of peanut and soybean allergens, respectively, after exposure
to enzymes mimicking human digestion [
110
]. The different outcome for major food allergen sources
was underlined by a study performed more than 10 years later using codfish as a model antigen [
111
].
After digestion with simulated gastric fluid, the IgE binding capacity of codfish proteins was reduced
more than 10,000-fold. This was shown in a reduced histamine release activity from basophil of
healthy donors, which were passively sensitized with sera from codfish allergic patients. Also in a
clinical setting, the impact of gastric enzymes on fish allergenicity was confirmed [
94
]. The diameter of
positive skin test reactions was significantly reduced after pre-digestion of allergens. Furthermore,
the lowest observed adverse effect level in double-blind, placebo controlled food challenges (DBPCFC)
was significantly higher. The pre-digestion was performed with gastric enzyme tablets clinically used
for patients with reduced gastric acid secretion. Also for celery allergens, the influence of gastric
enzymatic digestion on allergenicity could be confirmed in celery allergic patients with a mean age
of 72 years [
112
]. Even in this age group, skin test reactivity was significantly altered when test
allergens were pre-incubated with digestive enzymes, highlighting the impact of gastric digestion on
food allergenicity.
Deduced from these data, enzymatic hydrolysis of food proteins could help to reduce the
IgE-binding capacity and allergenicity in allergic patients. In our group, we could show that insects,
which are used as novel food, can be treated with enzymes from the food industry for protein
breakdown. The remaining smaller peptides or amino acids from the insect extracts completely lost
their cross-recognition of IgE from shrimp- and house dust mite-allergic patients and more important
also lost their capacity to elicit positive skin prick test reactions in shrimp-allergic patients [Pali-Schöll
et al., MS in revision].
Besides IgE binding, allergenicity has been additionally defined as the capacity of proteins to elicit
IgE formation [
113
]. Based on this definition, not only in situations with already establish food allergy,
but also during the development of food adverse reactions, protein degradation might play a major
role in the context of allergenicity. As mentioned above also for murine models, interference with
gastrointestinal digestion was confirmed to play a major role also in food allergy development. Most
studies evaluated situations of impaired gastric acid secretion due to anti-ulcer drug intake. In a first
study 152 adult patients being treated for 3 months with either H2-receptor or proton pump inhibitors
due to dyspeptic disorders such as reflux, gastritis erosions, or gastric ulcers, were screened for food
specific IgE reactivity. A boost of existing IgE or de novo IgE formation was found in one fourth of
all included patients [
95
]. In a sub-group of these patients who had developed hazelnut-specific IgE
during anti-ulcer treatment, not only could sensitization towards hazelnut be confirmed by specific IgE
antibodies and positive skin prick tests: hazelnut allergy was proved in 3 out of 5 patients with elevated
hazelnut-specific IgE titers after the 3 months treatment with gastric acid-suppression medication
also by positive provocation tests [
93
]. Moreover, for aged patients living in a geriatric nursing home,
the intake of anti-ulcer drugs was found to be associated with a significant shift of the immune response
towards a type 2-environment [
114
]. Not only in elderly, but also in pediatric patients, anti-ulcer drug
intake was reported to be associated with the development of food allergy [
115
,
116
]. In line, a recent
cohort study of 792,130 children demonstrated a higher allergy risk for children being treated with
either antibiotics or acid-suppressive medication during the first 6 months of life [7].
Importantly, this influence factor of hindered gastric digestion also seems to play a role during
pregnancy, where anti-acid medication of the mother leads to an enhanced risk of asthma or allergy in
the offspring in the mouse model [
104
]. In humans large health register studies and meta-analyses
confirmed the increased risk associated with intake of this medication during pregnancy for the
development of allergic diseases in children later in life [
117
120
], even though prospective studies
are missing.
Underlining the role of protein digestion during the sensitization process to food allergen, not only
hindrance of digestion due to gastric acid-suppressive medication, but also restriction of digestion
Nutrients 2018,10, 1129 8 of 16
due to bariatric gastric bypass surgery might play a fundamental role. To limit the caloric intake
of morbidly obese patients, only a small pouch of the stomach remains after surgery interventions
such as Roux-en-Y gastric bypass or sleeve gastrectomy [
121
]. In a pilot study sensitization to an
increasing number of common food compounds was detected after gastric bypass surgery [
122
]. These
studies highlight the important role of protein digestibility in the context of allergenicity. However,
it is obvious that protein digestion is one of the determinants influencing food allergenicity among
others, like protein solubility, size or abundance in a specific food [123].
Thus, it seems to be of special relevance to consider that impaired enzymatic protein digestion is
associated with enhanced allergenicity of food proteins. Different mechanisms may be of relevance:
(i) a hindered protein digestion through elevated gastric pH or reduced digestive capacity due to
bariatric surgery could result in bigger protein fragments that would be recognized by the cells of the
immune system; (ii) contained Th2-adjuvants (like aluminum in sucralfate) could direct the immune
response towards a Th2-response [
100
,
103
], and the allergic milieu could then even be transferred
from pregnant/lactating mothers to the offspring [
104
]; (iii) the dietary content changed during
acid-suppression with different remnants ending up in the lower digestive tract could change the
composition of the microbiome [98].
5. Summary
A number of factors influence the development of food allergies, including the situation in
the digestive system. An interference with proper digestion and absorption can be posed by
(i) food processing (Maillard reaction, aggregation) [
89
], (ii) suppression of gastric acid [
109
,
124
],
(iii) application of adjuvant substances into the digestive tract (aluminum components) [
100
,
103
],
or (iv) deletion of parts of the digestive system (bariatric surgery) [
122
]. Several of these processes and
factors have been shown to influence the digestive process
in vitro
, and for some of them the
in vivo
effect on allergenicity was proven (like for anti-acid drugs). Nevertheless, many knowledge gaps still
exist with need for further research studies (see Box 1).
As many of these factors came into play only recently in human evolution, they could probably
also explain an important part of the recent increase in prevalence of adverse food reactions.
Box 1. Knowledge gaps.
What Is Well Established? What Should be Further Investigated?
Amylase action influences the resulting remnants of
ingested starch and thereby the microbiome
Whether different amylase action and concentration,
e.g., in stress situations, also leads to different outcome
regarding allergenicity of the food
Food processing changes protein structure
and digestibility
Whether food processing might impact on gastrointestinal
pH levels
Heating can lead to glycation and Maillard products,
and thereby influences digestibility of involved proteins
Whether proteins become more able to sensitize, or to elicit
reactions in allergic patients
Anti-ulcer medication and antacids elevate gastric pH
levels and consequently influence food protein digestion
Whether gastric acid suppression influences also intestinal
pH levels and small intestinal protein digestion
Loading of lipid transfer proteins (LTP) with ligands
changes their digestibility
Whether loading of LTP changes their immunogenicity and
allergenicity in vivo
Blocking of gastric digestion increases the risk for allergic
sensitization
Whether the subsequent intestinal digestion is also
influenced by the changed gastric pH.
Whether a functional intestinal digestion could equalize the
detrimental sensitizing effect of a blocked gastric digestion
Author Contributions:
I.P.-S. contributed the part on digestibility of proteins and animal models of gastric acid
inhibition; E.U. added the part on the human data for acid-suppressing drugs and bariatric surgery; E.J.-J. wrote
the text about amylase action; M.K. performed the
in vitro
experiments and wrote the methods part for the figure.
All authors have seen and approved the final version of the manuscript.
Funding:
Research during this review was supported by the Austrian Science Fund FWF (grants SFB F4606-B28
to E.J.-J. and KLI284-B00 to E.U.) and a research grant of Nordmark Arzneimittel GmbH & Co. KG (to E.U.).
Conflicts of Interest: The authors declare no conflict of interest.
Nutrients 2018,10, 1129 9 of 16
Abbreviations
AGE advanced glycated end products
ATI amylase trypsin inhibitor
DBPCFC double-blind placebo-controlled food challenge
GIT gastrointestinal tract
MR Maillard reaction
MRP Maillard reaction products
NCGS non-celiac gluten-sensitivity
OVA ovalbumin
PC phosphatidylcholine
PPI proton pump inhibitor
RDS rapidly-digestible starch
RS resistant starch
SDS slowly-digestible starch
References
1.
U.S. Department of Health and Human Services. National Health Interview Survey. Available online:
https://ftp.cdc.gov/pub/Health_Statistics/NCHS/NHIS/SHS/2016_SHS_Table_C-2.pdf (accessed on
27 June 2018).
2.
Saulyte, J.; Regueira, C.; Montes-Martinez, A.; Khudyakov, P.; Takkouche, B. Active or passive exposure
to tobacco smoking and allergic rhinitis, allergic dermatitis, and food allergy in adults and children:
A systematic review and meta-analysis. PLoS Med. 2014,11, e1001611. [CrossRef] [PubMed]
3.
Untersmayr, E.; Diesner, S.C.; Oostingh, G.J.; Selzle, K.; Pfaller, T.; Schultz, C.; Zhang, Y.; Krishnamurthy, D.;
Starkl, P.; Knittelfelder, R.; et al. Nitration of the egg-allergen ovalbumin enhances protein allergenicity
but reduces the risk for oral sensitization in a murine model of food allergy. PLoS ONE
2010
,5, e14210.
[CrossRef] [PubMed]
4.
Allen, K.J.; Koplin, J.J.; Ponsonby, A.L.; Gurrin, L.C.; Wake, M.; Vuillermin, P.; Martin, P.; Matheson, M.;
Lowe, A.; Robinson, M.; et al. Vitamin d insufficiency is associated with challenge-proven food allergy in
infants. J. Allergy Clin. Immunol. 2013,131, 1109–1116. [CrossRef] [PubMed]
5.
Du Toit, G.; Sampson, H.A.; Plaut, M.; Burks, A.W.; Akdis, C.A.; Lack, G. Food allergy: Update on prevention
and tolerance. J. Allergy Clin. Immunol. 2018,141, 30–40. [CrossRef] [PubMed]
6.
Michel, S.; Busato, F.; Genuneit, J.; Pekkanen, J.; Dalphin, J.C.; Riedler, J.; Mazaleyrat, N.; Weber, J.;
Karvonen, A.M.; Hirvonen, M.R.; et al. Farm exposure and time trends in early childhood may influence
DNA methylation in genes related to asthma and allergy. Allergy 2013,68, 355–364. [CrossRef] [PubMed]
7.
Mitre, E.; Susi, A.; Kropp, L.E.; Schwartz, D.J.; Gorman, G.H.; Nylund, C.M. Association between use
of acid-suppressive medications and antibiotics during infancy and allergic diseases in early childhood.
JAMA Pediatr. 2018,172, e180315. [CrossRef] [PubMed]
8.
Jeurink, P.V.; Knipping, K.; Wiens, F.; Baranska, K.; Stahl, B.; Garssen, J.; Krolak-Olejnik, B. Importance of
maternal diet in the training of the infant’s immune system during gestation and lactation. Crit. Rev. Food
Sci. Nutr. 2018. [CrossRef] [PubMed]
9.
West, C. Introduction of complementary foods to infants. Ann. Nutr. Metab.
2017
,70 (Suppl. S2), 47–54.
[CrossRef] [PubMed]
10.
Fewtrell, M.; Bronsky, J.; Campoy, C.; Domellof, M.; Embleton, N.; Fidler Mis, N.; Hojsak, I.; Hulst, J.M.;
Indrio, F.; Lapillonne, A.; et al. Complementary feeding: A position paper by the european society
for paediatric gastroenterology, hepatology, and nutrition (espghan) committee on nutrition. J. Pediatr.
Gastroenterol. Nutr. 2017,64, 119–132. [CrossRef] [PubMed]
11.
Du Toit, G.; Katz, Y.; Sasieni, P.; Mesher, D.; Maleki, S.J.; Fisher, H.R.; Fox, A.T.; Turcanu, V.; Amir, T.;
Zadik-Mnuhin, G.; et al. Early consumption of peanuts in infancy is associated with a low prevalence of
peanut allergy. J. Allergy Clin. Immunol. 2008,122, 984–991. [CrossRef] [PubMed]
12.
Fisher, H.; Toit, G.D.; Bahnson, H.T.; Lack, G. The challenges of preventing food allergy: Lessons learned
from leap and eat. Ann. Allergy Asthma Immunol. 2018, in press.
Nutrients 2018,10, 1129 10 of 16
13.
du Toit, G.; Sayre, P.H.; Roberts, G.; Lawson, K.; Sever, M.L.; Bahnson, H.T.; Fisher, H.R.; Feeney, M.;
Radulovic, S.; Basting, M.; et al. Allergen specificity of early peanut consumption and effect on development
of allergic disease in the learning early about peanut allergy study cohort. J. Allergy Clin. Immunol.
2018
,141,
1343–1353. [CrossRef] [PubMed]
14.
Lin, A.H. Structure and digestion of common complementary food starches. J. Pediatr. Gastroenterol. Nutr.
2018,66, S35–S38. [CrossRef] [PubMed]
15.
Taniguchi, H.; Honnda, Y. Amylases. In Encyclopedia of Microbiology, 3rd ed.; Schaechter, M., Ed.; Academic
Press: Cambridge, MA, USA, 2009; pp. 159–173.
16.
Shulman, R.J. Starch malabsorption in infants. J. Pediatr. Gastroenterol. Nutr.
2018
,66 (Suppl. S3), S65–S67.
[CrossRef] [PubMed]
17.
Pimentel, A.C.; Barroso, I.G.; Ferreira, J.M.J.; Dias, R.O.; Ferreira, C.; Terra, W.R. Molecular machinery
of starch digestion and glucose absorption along the midgut of musca domestica. J. Insect Physiol.
2018
,
109, 11–20. [CrossRef] [PubMed]
18.
Pierzynowska, K.; Valverde-Piedra, J.; Szymanczyk, S.; Prykhod’ko, O.; Pieszka, M.; Kardas, M.;
Grochowska-Niedworok, E.; Grabowski, T.; Winiarczyk, M.; Pierzynowski, S. Pancreatic-like enzymes
of microbial origin restore growth and normalize lipid absorption in a pig model with exocrine pancreatic
insufficiency. Arch. Med. Sci. 2018,14, 407–414. [CrossRef] [PubMed]
19.
Meijster, T.; Tielemans, E.; Heederik, D. Effect of an intervention aimed at reducing the risk of allergic
respiratory disease in bakers: Change in flour dust and fungal alpha-amylase levels. Occup. Environ. Med.
2009,66, 543–549. [CrossRef] [PubMed]
20.
Peyrot des Gachons, C.; Breslin, P.A. Salivary amylase: Digestion and metabolic syndrome.
Curr. Diabetes Rep.
2016,16, 102. [CrossRef] [PubMed]
21.
Freitas, D.; Le Feunteun, S.; Panouille, M.; Souchon, I. The important role of salivary alpha-amylase in the
gastric digestion of wheat bread starch. Food Funct. 2018,9, 200–208. [CrossRef] [PubMed]
22.
Nater, U.M.; Rohleder, N. Salivary alpha-amylase as a non-invasive biomarker for the sympathetic nervous
system: Current state of research. Psychoneuroendocrinology 2009,34, 486–496. [CrossRef] [PubMed]
23.
Nater, U.M.; Skoluda, N.; Strahler, J. Biomarkers of stress in behavioural medicine. Curr. Opin. Psychiatry
2013,26, 440–445. [CrossRef] [PubMed]
24.
Bobel, T.S.; Hackl, S.B.; Langgartner, D.; Jarczok, M.N.; Rohleder, N.; Rook, G.A.; Lowry, C.A.; Gundel, H.;
Waller, C.; Reber, S.O. Less immune activation following social stress in rural vs. Urban participants raised
with regular or no animal contact, respectively. Proc. Natl. Acad. Sci. USA
2018
,115, 5259–5264. [CrossRef]
[PubMed]
25.
Dhital, S.; Warren, F.J.; Butterworth, P.J.; Ellis, P.R.; Gidley, M.J. Mechanisms of starch digestion by
alpha-amylase-structural basis for kinetic properties. Crit. Rev. Food Sci. Nutr.
2017
,57, 875–892. [CrossRef]
[PubMed]
26.
Dhital, S.; Lin, A.H.; Hamaker, B.R.; Gidley, M.J.; Muniandy, A. Mammalian mucosal alpha-glucosidases
coordinate with alpha-amylase in the initial starch hydrolysis stage to have a role in starch digestion beyond
glucogenesis. PLoS ONE 2013,8, e62546. [CrossRef] [PubMed]
27.
Lapis, T.J.; Penner, M.H.; Balto, A.S.; Lim, J. Oral digestion and perception of starch: Effects of cooking,
tasting time, and salivary alpha-amylase activity. Chem. Senses 2017,42, 635–645. [CrossRef] [PubMed]
28.
Takahama, U.; Hirota, S. Interactions of flavonoids with alpha-amylase and starch slowing down its digestion.
Food Funct. 2018,9, 677–687. [CrossRef] [PubMed]
29.
Farooq, A.M.; Dhital, S.; Li, C.; Zhang, B.; Huang, Q. Effects of palm oil on structural and
in vitro
digestion
properties of cooked rice starches. Int. J. Biol. Macromol. 2018,107, 1080–1085. [CrossRef] [PubMed]
30.
Martin, H.; Cordiner, S.B.; McGhie, T.K. Kiwifruit actinidin digests salivary amylase but not gastric lipase.
Food Funct. 2017,8, 3339–3345. [CrossRef] [PubMed]
31.
Smith, F.; Pan, X.; Bellido, V.; Toole, G.A.; Gates, F.K.; Wickham, M.S.; Shewry, P.R.; Bakalis, S.; Padfield, P.;
Mills, E.N. Digestibility of gluten proteins is reduced by baking and enhanced by starch digestion. Mol. Nutr.
Food Res. 2015,59, 2034–2043. [CrossRef] [PubMed]
32.
Schuppan, D.; Pickert, G.; Ashfaq-Khan, M.; Zevallos, V. Non-celiac wheat sensitivity: Differential diagnosis,
triggers and implications. Best Pract. Res. Clin. Gastroenterol. 2015,29, 469–476. [CrossRef] [PubMed]
Nutrients 2018,10, 1129 11 of 16
33.
Bellinghausen, I.; Weigmann, B.; Zevallos, V.; Maxeiner, J.; Reissig, S.; Waisman, A.; Schuppan, D.; Saloga, J.
Wheat amylase-trypsin inhibitors exacerbate intestinal and airway allergic immune responses in humanized
mice. J. Allergy Clin. Immunol. 2018. [CrossRef] [PubMed]
34.
Zevallos, V.F.; Raker, V.K.; Maxeiner, J.; Scholtes, P.; Steinbrink, K.; Schuppan, D. Dietary wheat amylase
trypsin inhibitors exacerbate murine allergic airway inflammation. Eur. J. Nutr.
2018
. [CrossRef] [PubMed]
35.
Fardet, A. Wheat-based foods and non celiac gluten/wheat sensitivity: Is drastic processing the main key
issue? Med. Hypotheses 2015,85, 934–939. [CrossRef] [PubMed]
36.
Herrmann, E.; Young, W.; Reichert-Grimm, V.; Weis, S.; Riedel, C.U.; Rosendale, D.; Stoklosinski, H.; Hunt, M.;
Egert, M.
In vivo
assessment of resistant starch degradation by the caecal microbiota of mice using rna-based
stable isotope probing—A proof-of-principle study. Nutrients 2018,10, 179. [CrossRef] [PubMed]
37.
Bello-Perez, L.A.; Flores-Silva, P.C.; Agama-Acevedo, E.; Tovar, J. Starch digestibility: Past, present,
and future. J. Sci. Food Agric. 2018. [CrossRef] [PubMed]
38.
Warren, F.J.; Fukuma, N.M.; Mikkelsen, D.; Flanagan, B.M.; Williams, B.A.; Lisle, A.T.; Ó’Cuív, P.;
Morrison, M.; Gidley, M.J. Food starch structure impacts gut microbiome composition. mSphere
2018
,
3, e00086-18. [CrossRef] [PubMed]
39.
Newman, M.A.; Petri, R.M.; Grull, D.; Zebeli, Q.; Metzler-Zebeli, B.U. Transglycosylated starch modulates
the gut microbiome and expression of genes related to lipid synthesis in liver and adipose tissue of pigs.
Front. Microbiol. 2018,9, 224. [CrossRef] [PubMed]
40.
Maier, T.V.; Lucio, M.; Lee, L.H.; VerBerkmoes, N.C.; Brislawn, C.J.; Bernhardt, J.; Lamendella, R.;
McDermott, J.E.; Bergeron, N.; Heinzmann, S.S.; et al. Impact of dietary resistant starch on the human gut
microbiome, metaproteome, and metabolome. mBio 2017,8, e01343-17. [CrossRef] [PubMed]
41.
Stefka, A.T.; Feehley, T.; Tripathi, P.; Qiu, J.; McCoy, K.; Mazmanian, S.K.; Tjota, M.Y.; Seo, G.Y.; Cao, S.;
Theriault, B.R.; et al. Commensal bacteria protect against food allergen sensitization. Proc. Natl. Acad.
Sci. USA 2014,111, 13145–13150. [CrossRef] [PubMed]
42.
Ho, H.E.; Bunyavanich, S. Role of the microbiome in food allergy. Curr. Allergy Asthma Rep.
2018
,18, 27.
[CrossRef] [PubMed]
43.
Huang, Y.J.; Marsland, B.J.; Bunyavanich, S.; O’Mahony, L.; Leung, D.Y.; Muraro, A.; Fleisher, T.A.
The microbiome in allergic disease: Current understanding and future opportunities-2017 practall document
of the American academy of allergy, asthma & immunology and the European academy of allergy and
clinical immunology. J. Allergy Clin. Immunol. 2017,139, 1099–1110. [PubMed]
44.
Bogh, K.L.; Barkholt, V.; Madsen, C.B. The sensitising capacity of intact beta-lactoglobulin is reduced by
co-administration with digested beta-lactoglobulin. Int. Arch. Allergy Immunol.
2013
,161, 21–36. [CrossRef]
[PubMed]
45.
Berecz, B.; Clare Mills, E.N.; Paradi, I.; Lang, F.; Tamas, L.; Shewry, P.R.; Mackie, A.R. Stability of sunflower
2s albumins and ltp to physiologically relevant
in vitro
gastrointestinal digestion. Food Chem.
2013
,138,
2374–2381. [CrossRef] [PubMed]
46.
Bartra, J.; Garcia-Moral, A.; Enrique, E. Geographical differences in food allergy. Bundesgesundheitsblatt
Gesundheitsforsch. Gesundheitssch. 2016,59, 755–763. [CrossRef] [PubMed]
47.
Moreno, F.J.; Mackie, A.R.; Mills, E.N. Phospholipid interactions protect the milk allergen alpha-lactalbumin
from proteolysis during in vitro digestion. J. Agric. Food Chem. 2005,53, 9810–9816. [CrossRef] [PubMed]
48.
Mandalari, G.; Adel-Patient, K.; Barkholt, V.; Baro, C.; Bennett, L.; Bublin, M.; Gaier, S.; Graser, G.; Ladics, G.S.;
Mierzejewska, D.; et al.
In vitro
digestibility of beta-casein and beta-lactoglobulin under simulated human
gastric and duodenal conditions: A multi-laboratory evaluation. Regul. Toxicol. Pharmacol.
2009
,55, 372–381.
[CrossRef] [PubMed]
49.
Abdullah, S.U.; Alexeev, Y.; Johnson, P.E.; Rigby, N.M.; Mackie, A.R.; Dhaliwal, B.; Mills, E.N. Ligand
binding to an allergenic lipid transfer protein enhances conformational flexibility resulting in an increase in
susceptibility to gastroduodenal proteolysis. Sci. Rep. 2016,6, 30279. [CrossRef] [PubMed]
50.
Pastorello, E.A.; Farioli, L.; Conti, A.; Pravettoni, V.; Bonomi, S.; Iametti, S.; Fortunato, D.; Scibilia, J.;
Bindslev-Jensen, C.; Ballmer-Weber, B.; et al. Wheat ige-mediated food allergy in european patients:
Alpha-amylase inhibitors, lipid transfer proteins and low-molecular-weight glutenins. Allergenic molecules
recognized by double-blind, placebo-controlled food challenge. Int. Arch. Allergy Immunol.
2007
,144, 10–22.
[CrossRef] [PubMed]
Nutrients 2018,10, 1129 12 of 16
51.
Palacin, A.; Quirce, S.; Armentia, A.; Fernandez-Nieto, M.; Pacios, L.F.; Asensio, T.; Sastre, J.; Diaz-Perales, A.;
Salcedo, G. Wheat lipid transfer protein is a major allergen associated with baker’s asthma. J. Allergy
Clin. Immunol. 2007,120, 1132–1138. [CrossRef] [PubMed]
52.
Di Stasio, L.; Picariello, G.; Mongiello, M.; Nocerino, R.; Berni Canani, R.; Bavaro, S.; Monaci, L.; Ferranti, P.;
Mamone, G. Peanut digestome: Identification of digestion resistant ige binding peptides. Food Chem. Toxicol.
2017,107, 88–98. [CrossRef] [PubMed]
53.
Verhoeckx, K.C.; Vissers, Y.M.; Baumert, J.L.; Faludi, R.; Feys, M.; Flanagan, S.; Herouet-Guicheney, C.;
Holzhauser, T.; Shimojo, R.; van der Bolt, N.; et al. Food processing and allergenicity. Food Chem. Toxicol.
2015,80, 223–240. [CrossRef] [PubMed]
54.
Roth-Walter, F.; Berin, M.C.; Arnaboldi, P.; Escalante, C.R.; Dahan, S.; Rauch, J.; Jensen-Jarolim, E.; Mayer, L.
Pasteurization of milk proteins promotes allergic sensitization by enhancing uptake through peyer’s patches.
Allergy 2008,63, 882–890. [CrossRef] [PubMed]
55.
Luz Sanz, M.; Corzo-Martinez, M.; Rastall, R.A.; Olano, A.; Moreno, F.J. Characterization and
in vitro
digestibility of bovine beta-lactoglobulin glycated with galactooligosaccharides. J. Agric. Food Chem.
2007
,
55, 7916–7925. [CrossRef] [PubMed]
56.
Seo, S.; L’Hocine, L.; Karboune, S. Allergenicity of potato proteins and of their conjugates with galactose,
galactooligosaccharides, and galactan in native, heated, and digested forms. J. Agric. Food Chem.
2014
,62,
3591–3598. [CrossRef] [PubMed]
57.
Nakamura, A.; Watanabe, K.; Ojima, T.; Ahn, D.H.; Saeki, H. Effect of maillard reaction on allergenicity of
scallop tropomyosin. J. Agric. Food Chem. 2005,53, 7559–7564. [CrossRef] [PubMed]
58.
Maleki, S.J.; Schmitt, D.A.; Galeano, M.; Hurlburt, B.K. Comparison of the digestibility of the major peanut
allergens in thermally processed peanuts and in pure form. Foods 2014,3, 290–303. [CrossRef] [PubMed]
59.
Simonato, B.; Pasini, G.; Giannattasio, M.; Peruffo, A.D.; De Lazzari, F.; Curioni, A. Food allergy to wheat
products: The effect of bread baking and
in vitro
digestion on wheat allergenic proteins. A study with bread
dough, crumb, and crust. J. Agric. Food Chem. 2001,49, 5668–5673. [CrossRef] [PubMed]
60.
Jimenez-Saiz, R.; Belloque, J.; Molina, E.; Lopez-Fandino, R. Human immunoglobulin e (ige) binding to
heated and glycated ovalbumin and ovomucoid before and after
in vitro
digestion. J. Agric. Food Chem.
2011
,
59, 10044–10051. [CrossRef] [PubMed]
61.
Downs, M.L.; Baumert, J.L.; Taylor, S.L.; Mills, E.N. Mass spectrometric analysis of allergens in roasted
walnuts. J. Proteomics 2016,142, 62–69. [CrossRef] [PubMed]
62.
Yeboah, F.K.; Alli, I.; Yaylayan, V.A.; Yasuo, K.; Chowdhury, S.F.; Purisima, E.O. Effect of limited solid-state
glycation on the conformation of lysozyme by esi-msms peptide mapping and molecular modeling.
Bioconjug. Chem. 2004,15, 27–34. [CrossRef] [PubMed]
63.
De Jongh, H.H.; Taylor, S.L.; Koppelman, S.J. Controlling the aggregation propensity and thereby digestibility
of allergens by maillardation as illustrated for cod fish parvalbumin. J. Biosci. Bioeng.
2011
,111, 204–211.
[CrossRef] [PubMed]
64.
Corzo-Martinez, M.; Soria, A.C.; Belloque, J.; Villamiel, M.; Moreno, F.J. Effect of glycation on the
gastrointestinal digestibility and immunoreactivity of bovine b-lactoglobulin. Int. Dairy J.
2012
,20, 742–752.
[CrossRef]
65.
Scheijen, J.; Hanssen, N.M.J.; van Greevenbroek, M.M.; Van der Kallen, C.J.; Feskens, E.J.M.;
Stehouwer, C.D.A.; Schalkwijk, C.G. Dietary intake of advanced glycation endproducts is associated with
higher levels of advanced glycation endproducts in plasma and urine: The codam study. Clin. Nutr.
2018
,37,
919–925. [CrossRef] [PubMed]
66.
Hellwig, M.; Bunzel, D.; Huch, M.; Franz, C.M.; Kulling, S.E.; Henle, T. Stability of individual maillard
reaction products in the presence of the human colonic microbiota. J. Agric. Food Chem.
2015
,63, 6723–6730.
[CrossRef] [PubMed]
67.
Gupta, R.K.; Gupta, K.; Sharma, A.; Das, M.; Ansari, I.A.; Dwivedi, P.D. Maillard reaction in food allergy:
Pros and cons. Crit. Rev. Food Sci. Nutr. 2018,58, 208–226. [CrossRef] [PubMed]
68.
Buttari, B.; Profumo, E.; Capozzi, A.; Facchiano, F.; Saso, L.; Sorice, M.; Rigano, R. Advanced glycation end
products of human beta(2) glycoprotein i modulate the maturation and function of dcs. Blood
2011
,117,
6152–6161. [CrossRef] [PubMed]
Nutrients 2018,10, 1129 13 of 16
69.
Moghaddam, A.E.; Hillson, W.R.; Noti, M.; Gartlan, K.H.; Johnson, S.; Thomas, B.; Artis, D.; Sattentau, Q.J.
Dry roasting enhances peanut-induced allergic sensitization across mucosal and cutaneous routes in mice.
J. Allergy Clin. Immunol. 2014,134, 1453–1456. [CrossRef] [PubMed]
70.
Mueller, G.A.; Maleki, S.J.; Johnson, K.; Hurlburt, B.K.; Cheng, H.; Ruan, S.; Nesbit, J.B.; Pomes, A.;
Edwards, L.L.; Schorzman, A.; et al. Identification of maillard reaction products on peanut allergens
that influence binding to the receptor for advanced glycation end products. Allergy
2013
,68, 1546–1554.
[CrossRef] [PubMed]
71.
Cabanillas, B.; Maleki, S.J.; Cheng, H.; Novak, N. Differences in the uptake of ara h 3 from raw and roasted
peanut by monocyte-derived dendritic cells. Int. Arch. Allergy Immunol. 2018. [CrossRef] [PubMed]
72.
Hilmenyuk, T.; Bellinghausen, I.; Heydenreich, B.; Ilchmann, A.; Toda, M.; Grabbe, S.; Saloga, J. Effects of
glycation of the model food allergen ovalbumin on antigen uptake and presentation by human dendritic
cells. Immunology 2010,129, 437–445. [CrossRef] [PubMed]
73.
Ilchmann, A.; Burgdorf, S.; Scheurer, S.; Waibler, Z.; Nagai, R.; Wellner, A.; Yamamoto, Y.; Yamamoto, H.;
Henle, T.; Kurts, C.; et al. Glycation of a food allergen by the maillard reaction enhances its t-cell
immunogenicity: Role of macrophage scavenger receptor class a type I and II. J. Allergy Clin. Immunol.
2010
,
125, 175–183. [CrossRef] [PubMed]
74.
Toda, M.; Heilmann, M.; Ilchmann, A.; Vieths, S. The maillard reaction and food allergies: Is there a link?
Clin. Chem. Lab. Med. 2014,52, 61–67. [CrossRef] [PubMed]
75.
Seiquer, I.; Diaz-Alguacil, J.; Delgado-Andrade, C.; Lopez-Frias, M.; Munoz Hoyos, A.; Galdo, G.;
Navarro, M.P. Diets rich in maillard reaction products affect protein digestibility in adolescent males
aged 11–14 y. Am. J. Clin. Nutr. 2006,83, 1082–1088. [CrossRef] [PubMed]
76.
Heilmann, M.; Wellner, A.; Gadermaier, G.; Ilchmann, A.; Briza, P.; Krause, M.; Nagai, R.; Burgdorf, S.;
Scheurer, S.; Vieths, S.; et al. Ovalbumin modified with pyrraline, a maillard reaction product, shows
enhanced t-cell immunogenicity. J. Biol. Chem. 2014,289, 7919–7928. [CrossRef] [PubMed]
77.
Han, X.Y.; Yang, H.; Rao, S.T.; Liu, G.Y.; Hu, M.J.; Zeng, B.C.; Cao, M.J.; Liu, G.M. The maillard reaction
reduced the sensitization of tropomyosin and arginine kinase from scylla paramamosain, simultaneously.
J. Agric. Food Chem. 2018,66, 2934–2943. [CrossRef] [PubMed]
78.
Suzuki, Y.; Kassai, M.; Hirose, T.; Katayama, S.; Nakamura, K.; Akiyama, H.; Teshima, R.; Nakamura, S.
Modulation of immunoresponse in balb/c mice by oral administration of fag e 1-glucomannan conjugate.
J. Agric. Food Chem. 2009,57, 9787–9792. [CrossRef] [PubMed]
79.
Gupta, R.K.; Raghav, A.; Sharma, A.; Gupta, K.; Neelabh; Mandal, P.; Tripathi, A.; Ansari, I.A.; Das, M.;
Dwivedi, P.D. Glycation of clinically relevant chickpea allergen attenuates its allergic immune response in
balb/c mice. Food Chem. 2017,235, 244–256. [CrossRef] [PubMed]
80.
Rahaman, T.; Vasiljevic, T.; Ramchandran, L. Conformational changes of beta-lactoglobulin induced by shear,
heat, and ph-effects on antigenicity. J. Dairy Sci. 2015,98, 4255–4265. [CrossRef] [PubMed]
81.
Scheurer, S.; Lauer, I.; Foetisch, K.; San Miguel Moncin, M.; Retzek, M.; Hartz, C.; Enrique, E.; Lidholm, J.;
Cistero-Bahima, A.; Vieths, S. Strong allergenicity of pru av 3, the lipid transfer protein from cherry, is related
to high stability against thermal processing and digestion. J. Allergy Clin. Immunol.
2004
,114, 900–907.
[CrossRef] [PubMed]
82.
Malanin, K.; Lundberg, M.; Johansson, S.G. Anaphylactic reaction caused by neoallergens in heated pecan
nut. Allergy 1995,50, 988–991. [CrossRef] [PubMed]
83.
Codina, R.; Oehling, A.G., Jr.; Lockey, R.F. Neoallergens in heated soybean hull. Int. Arch. Allergy Immunol.
1998,117, 120–125. [CrossRef] [PubMed]
84.
Gruber, P.; Vieths, S.; Wangorsch, A.; Nerkamp, J.; Hofmann, T. Maillard reaction and enzymatic browning
affect the allergenicity of pru av 1, the major allergen from cherry (prunus avium). J. Agric. Food Chem.
2004
,
52, 4002–4007. [CrossRef] [PubMed]
85.
Cucu, T.; De Meulenaer, B.; Bridts, C.; Devreese, B.; Ebo, D. Impact of thermal processing and the maillard
reaction on the basophil activation of hazelnut allergic patients. Food Chem. Toxicol.
2012
,50, 1722–1728.
[CrossRef] [PubMed]
86.
Xu, L.; Gong, Y.; Gern, J.E.; Ikeda, S.; Lucey, J.A. Glycation of whey protein with dextrans of different molar
mass: Effect on immunoglobulin e-binding capacity with blood sera obtained from patients with cow milk
protein allergy. J. Dairy Sci. 2018,101, 6823–6834. [CrossRef] [PubMed]
Nutrients 2018,10, 1129 14 of 16
87.
Maleki, S.J.; Chung, S.Y.; Champagne, E.T.; Raufman, J.P. The effects of roasting on the allergenic properties
of peanut proteins. J. Allergy Clin. Immunol. 2000,106, 763–768. [CrossRef] [PubMed]
88.
Hansen, K.S.; Ballmer-Weber, B.K.; Luttkopf, D.; Skov, P.S.; Wuthrich, B.; Bindslev-Jensen, C.; Vieths, S.;
Poulsen, L.K. Roasted hazelnuts—Allergenic activity evaluated by double-blind, placebo-controlled food
challenge. Allergy 2003,58, 132–138. [CrossRef] [PubMed]
89.
Teodorowicz, M.; van Neerven, J.; Savelkoul, H. Food processing: The influence of the maillard reaction on
immunogenicity and allergenicity of food proteins. Nutrients 2017,9, 835. [CrossRef] [PubMed]
90.
Hantusch, B.; Knittelfelder, R.; Wallmann, J.; Krieger, S.; Szalai, K.; Untersmayr, E.; Vogel, M.; Stadler, B.M.;
Scheiner, O.; Boltz-Nitulescu, G.; et al. Internal images: Human anti-idiotypic fab antibodies mimic the ige
epitopes of grass pollen allergen phl p 5a. Mol. Immunol. 2006,43, 2180–2187. [CrossRef] [PubMed]
91.
Ortolani, C.; Ballmer-Weber, B.K.; Hansen, K.S.; Ispano, M.; Wuthrich, B.; Bindslev-Jensen, C.; Ansaloni, R.;
Vannucci, L.; Pravettoni, V.; Scibilia, J.; et al. Hazelnut allergy: A double-blind, placebo-controlled food
challenge multicenter study. J. Allergy Clin. Immunol. 2000,105, 577–581. [CrossRef] [PubMed]
92.
Herman, R.; Gao, Y.; Storer, N. Acid-induced unfolding kinetics in simulated gastric digestion of proteins.
Regul. Toxicol. Pharmacol. 2006,46, 93–99. [CrossRef] [PubMed]
93.
Schöll, I.; Untersmayr, E.; Bakos, N.; Roth-Walter, F.; Gleiss, A.; Boltz-Nitulescu, G.; Scheiner, O.;
Jensen-Jarolim, E. Antiulcer drugs promote oral sensitization and hypersensitivity to hazelnut allergens in
balb/c mice and humans. Am. J. Clin. Nutr. 2005,81, 154–160. [CrossRef] [PubMed]
94.
Untersmayr, E.; Vestergaard, H.; Malling, H.J.; Jensen, L.B.; Platzer, M.H.; Boltz-Nitulescu, G.; Scheiner, O.;
Skov, P.S.; Jensen-Jarolim, E.; Poulsen, L.K. Incomplete digestion of codfish represents a risk factor for
anaphylaxis in patients with allergy. J. Allergy Clin. Immunol. 2007,119, 711–717. [CrossRef] [PubMed]
95.
Untersmayr, E.; Bakos, N.; Scholl, I.; Kundi, M.; Roth-Walter, F.; Szalai, K.; Riemer, A.B.; Ankersmit, H.J.;
Scheiner, O.; Boltz-Nitulescu, G.; et al. Anti-ulcer drugs promote ige formation toward dietary antigens in
adult patients. FASEB J. 2005,19, 656–658. [CrossRef] [PubMed]
96.
Gardner, J.D.; Ciociola, A.A.; Robinson, M. Measurement of meal-stimulated gastric acid secretion by
in vivo
gastric autotitration. J. Appl. Physiol. 2002,92, 427–434. [CrossRef] [PubMed]
97.
Prichard, P.J.; Yeomans, N.D.; Mihaly, G.W.; Jones, D.B.; Buckle, P.J.; Smallwood, R.A.; Louis, W.J. Omeprazole:
A study of its inhibition of gastric ph and oral pharmacokinetics after morning or evening dosage.
Gastroenterology 1985,88, 64–69. [CrossRef]
98.
Diesner, S.C.; Bergmayr, C.; Pfitzner, B.; Assmann, V.; Krishnamurthy, D.; Starkl, P.; Endesfelder, D.;
Rothballer, M.; Welzl, G.; Rattei, T.; et al. A distinct microbiota composition is associated with protection from
food allergy in an oral mouse immunization model. Clin. Immunol. 2016,173, 10–18. [CrossRef] [PubMed]
99.
Riemer, A.B.; Gruber, S.; Pali-Scholl, I.; Kinaciyan, T.; Untersmayr, E.; Jensen-Jarolim, E. Suppression of
gastric acid increases the risk of developing immunoglobulin e-mediated drug hypersensitivity: Human
diclofenac sensitization and a murine sensitization model. Clin. Exp. Allergy
2010
,40, 486–493. [CrossRef]
[PubMed]
100.
Brunner, R.; Wallmann, J.; Szalai, K.; Karagiannis, P.; Altmeppen, H.; Riemer, A.B.; Jensen-Jarolim, E.;
Pali-Scholl, I. Aluminium per se and in the anti-acid drug sucralfate promotes sensitization via the oral route.
Allergy 2009,64, 890–897. [CrossRef] [PubMed]
101.
Diesner, S.C.; Knittelfelder, R.; Krishnamurthy, D.; Pali-Scholl, I.; Gajdzik, L.; Jensen-Jarolim, E.;
Untersmayr, E. Dose-dependent food allergy induction against ovalbumin under acid-suppression: A murine
food allergy model. Immunol. Lett. 2008,121, 45–51. [CrossRef] [PubMed]
102.
Pali-Scholl, I.; Yildirim, A.O.; Ackermann, U.; Knauer, T.; Becker, C.; Garn, H.; Renz, H.; Jensen-Jarolim, E.;
Fehrenbach, H. Anti-acids lead to immunological and morphological changes in the intestine of balb/c mice
similar to human food allergy. Exp. Toxicol. Pathol. 2008,60, 337–345. [CrossRef] [PubMed]
103.
Brunner, R.; Wallmann, J.; Szalai, K.; Karagiannis, P.; Kopp, T.; Scheiner, O.; Jensen-Jarolim, E.;
Pali-Scholl, I. The impact of aluminium in acid-suppressing drugs on the immune response of balb/c
mice. Clin. Exp. Allergy 2007,37, 1566–1573. [CrossRef] [PubMed]
104.
Scholl, I.; Ackermann, U.; Ozdemir, C.; Blumer, N.; Dicke, T.; Sel, S.; Sel, S.; Wegmann, M.; Szalai, K.;
Knittelfelder, R.; et al. Anti-ulcer treatment during pregnancy induces food allergy in mouse mothers and a
th2-bias in their offspring. FASEB J. 2007,21, 1264–1270. [CrossRef] [PubMed]
Nutrients 2018,10, 1129 15 of 16
105.
Ramirez, E.; Cabanas, R.; Laserna, L.S.; Fiandor, A.; Tong, H.; Prior, N.; Calderon, O.; Medrano, N.; Bobolea, I.;
Frias, J.; et al. Proton pump inhibitors are associated with hypersensitivity reactions to drugs in hospitalized
patients: A nested case-control in a retrospective cohort study. Clin. Exp. Allergy
2013
,43, 344–352. [CrossRef]
[PubMed]
106.
Pali-Scholl, I.; Herzog, R.; Wallmann, J.; Szalai, K.; Brunner, R.; Lukschal, A.; Karagiannis, P.; Diesner, S.C.;
Jensen-Jarolim, E. Antacids and dietary supplements with an influence on the gastric ph increase the risk for
food sensitization. Clin. Exp. Allergy 2010,40, 1091–1098. [CrossRef] [PubMed]
107.
Untersmayr, E.; Scholl, I.; Swoboda, I.; Beil, W.J.; Forster-Waldl, E.; Walter, F.; Riemer, A.; Kraml, G.;
Kinaciyan, T.; Spitzauer, S.; et al. Antacid medication inhibits digestion of dietary proteins and causes food
allergy: A fish allergy model in balb/c mice. J. Allergy Clin. Immunol. 2003,112, 616–623. [CrossRef]
108.
Untersmayr, E. Acid suppression therapy and allergic reactions. Allergo J. Int.
2015
,24, 303–311. [CrossRef]
[PubMed]
109.
Pali-Scholl, I.; Jensen-Jarolim, E. Anti-acid medication as a risk factor for food allergy. Allergy
2011
,66,
469–477. [CrossRef] [PubMed]
110.
Burks, A.W.; Williams, L.W.; Thresher, W.; Connaughton, C.; Cockrell, G.; Helm, R.M. Allergenicity of peanut
and soybean extracts altered by chemical or thermal denaturation in patients with atopic dermatitis and
positive food challenges. J. Allergy Clin. Immunol. 1992,90, 889–897. [CrossRef]
111.
Untersmayr, E.; Poulsen, L.K.; Platzer, M.H.; Pedersen, M.H.; Boltz-Nitulescu, G.; Skov, P.S.; Jensen-Jarolim, E.
The effects of gastric digestion on codfish allergenicity. J. Allergy Clin. Immunol.
2005
,115, 377–382. [CrossRef]
[PubMed]
112.
Untersmayr, E.; Diesner, S.C.; Bramswig, K.H.; Knittelfelder, R.; Bakos, N.; Gundacker, C.; Lukschal, A.;
Wallmann, J.; Szalai, K.; Pali-Scholl, I.; et al. Characterization of intrinsic and extrinsic risk factors for celery
allergy in immunosenescence. Mech. Ageing Dev. 2008,129, 120–128. [CrossRef] [PubMed]
113.
Aalberse, R.C. Structural biology of allergens. J. Allergy Clin. Immunol.
2000
,106, 228–238. [CrossRef]
[PubMed]
114.
Bakos, N.; Scholl, I.; Szalai, K.; Kundi, M.; Untersmayr, E.; Jensen-Jarolim, E. Risk assessment in elderly for
sensitization to food and respiratory allergens. Immunol. Lett. 2006,107, 15–21. [CrossRef] [PubMed]
115.
DeMuth, K.; Stecenko, A.; Sullivan, K.; Fitzpatrick, A. Relationship between treatment with antacid
medication and the prevalence of food allergy in children. Allergy Asthma Proc.
2013
,34, 227–232. [CrossRef]
[PubMed]
116.
Trikha, A.; Baillargeon, J.G.; Kuo, Y.F.; Tan, A.; Pierson, K.; Sharma, G.; Wilkinson, G.; Bonds, R.S.
Development of food allergies in patients with gastroesophageal reflux disease treated with gastric acid
suppressive medications. Pediatr. Allergy Immunol. 2013,24, 582–588. [CrossRef] [PubMed]
117.
Dehlink, E.; Yen, E.; Leichtner, A.M.; Hait, E.J.; Fiebiger, E. First evidence of a possible association between
gastric acid suppression during pregnancy and childhood asthma: A population-based register study.
Clin. Exp. Allergy 2009,39, 246–253. [CrossRef] [PubMed]
118.
Mulder, B.; Schuiling-Veninga, C.C.; Bos, H.J.; De Vries, T.W.; Jick, S.S.; Hak, E. Prenatal exposure to
acid-suppressive drugs and the risk of allergic diseases in the offspring: A cohort study. Clin. Exp. Allergy
2014,44, 261–269. [CrossRef] [PubMed]
119.
Lai, T.; Wu, M.; Liu, J.; Luo, M.; He, L.; Wang, X.; Wu, B.; Ying, S.; Chen, Z.; Li, W.; et al. Acid-suppressive drug
use during pregnancy and the risk of childhood asthma: A meta-analysis. Pediatrics
2018
,141, e20170889.
[CrossRef] [PubMed]
120.
Cea Soriano, L.; Hernandez-Diaz, S.; Johansson, S.; Nagy, P.; Garcia-Rodriguez, L.A. Exposure to
acid-suppressing drugs during pregnancy and the risk of asthma in childhood: An observational cohort
study. Aliment. Pharmacol. Ther. 2016,43, 427–437. [CrossRef] [PubMed]
121.
Kurian, M.; Kroh, M.; Chand, B.; Mikami, D.; Reavis, K.; Khaitan, L. Sages review of endoscopic and
minimally invasive bariatric interventions: A review of endoscopic and non-surgical bariatric interventions.
Surg. Endosc. 2018, in press. [CrossRef] [PubMed]
122.
Shakeri-Leidenmuhler, S.; Lukschal, A.; Schultz, C.; Bohdjalian, A.; Langer, F.; Birsan, T.; Diesner, S.C.;
Greisenegger, E.K.; Scheiner, O.; Kopp, T.; et al. Surgical elimination of the gastric digestion by roux-en-y
gastric bypass impacts on food sensitization—A pilot study. Obes. Surg.
2015
,25, 2268–2275. [CrossRef]
[PubMed]
Nutrients 2018,10, 1129 16 of 16
123.
Pekar, J.; Ret, D.; Untersmayr, E. Stability of allergens. Mol. Immunol.
2018
,100, 14–20. [CrossRef] [PubMed]
124.
Untersmayr, E.; Jensen-Jarolim, E. The role of protein digestibility and antacids on food allergy outcomes.
J. Allergy Clin. Immunol. 2008,121, 1301–1308. [CrossRef] [PubMed]
©
2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... All of these factors contribute to the great heterogeneity of protein digestion, with some proteins being extensively broken down into smaller peptides, while others pass through the gastrointestinal tract without being significantly degraded [30]. In the past, protein stability has been proposed as a measure of the immunogenic potential of different proteins, seeing as more stable proteins could preserve their conformational epitopes and elicit allergic responses. ...
... Nonetheless, phenolic compounds can also protect dietary protein from digestion, as is the case of wheat proteins treated with flavonoids extracted from onion skin [113]. This can prove a hindrance in the development of new therapeutic strategies, seeing as undigested proteins tend to keep their immunogenic potential [30]. Adding to this, an improved digestion of dietary proteins might not be enough to reduce their immunogenic potential, seeing as some large peptides formed during digestion maintain an ability to bind to IgEs [40]. ...
Article
Full-text available
Food allergies are becoming ever more prevalent around the world. This pathology is characterized by the breakdown of oral tolerance to ingested food allergens, resulting in allergic reactions in subsequent exposures. Due to the possible severity of the symptoms associated with this pathology, new approaches to prevent it and reduce associated symptoms are of utmost importance. In this framework, dietary phenolic compounds appear as a tool with a not fully explored potential. Some phenolic compounds have been pointed to with the ability to modulate food allergies and possibly reduce their symptoms. These compounds can modulate food allergies through many different mechanisms, such as altering the bioaccessibility and bioavailability of potentially immunogenic peptides, by modulating the human immune system and by modulating the composition of the human microbiome that resides in the oral cavity and the gastrointestinal tract. This review deepens the state-of-the-art of the modulation of these mechanisms by phenolic compounds. While this review shows clear evidence that dietary supplementation with foods rich in phenolic compounds might constitute a new approach to the management of food allergies, it also highlights the need for further research to delve into the mechanisms of action of these compounds and decipher systematic structure/activity relationships.
... Highly digestible proteins generally have their epitopes disintegrated more easily, reducing their potential to cause allergies [134]. Conversely, proteins that resist digestion can maintain their structure, including epitopes, and may cross the intestinal barrier to elicit an immune response, heightening allergy risks [106,119]. For instance, in the study conducted by Jin et al. [70], high-pressure processing highpressure processing led to the unfolding of the protein structure of the squid allergen Tod p1. ...
Article
Full-text available
Sustainable food practices within the food industry are pertinent to allow efficient food supply while not negatively impacting the environment. Alternative proteins have gained the attention of the food industry and consumers. To provide safe novel food products, these protein sources need to be assessed for potential allergen risk to ensure food safety and allow effective labelling to protect the consumer. In this review, the various detection assays applied to target potential allergens in novel and alternative foods are described together with their applications, mechanisms and limitations. Additionally, the use of non-thermal technologies to mitigate the reactivity of food allergens in these new products is explored. Non-thermal techniques including cold plasma, pulsed electric field, ultrasound and gamma irradiation are discussed. This review examines the potential mechanisms by which non-thermal technologies may reduce food allergenicity, primarily through alterations in protein epitopes that could affect antibody recognition. However, it is important to note that the understanding of the precise mechanisms and outcomes in allergen mitigation through these methods remains an area requiring further research.
... Numerous studies have demonstrated a correlation between increased digestibility and decreased allergenicity (Gasparini et al., 2022;Pali-Schöll et al., 2018;Wang et al., 2022). Consequently, serology experiments, an RBL-2H3 cell degranulation model, and a PCA mice model were conducted to evaluate the effect of traditional cooking methods on the allergenicity of egg proteins. ...
Article
Full-text available
Egg allergy is one of the most common food allergies globally. This study aimed to assess the impact of four traditional cooking methods on the allergenicity of egg proteins using a comprehensive strategy, including simulated gastrointestinal digestion in vitro, serology experiments, a rat basophilic leukemia (RBL)‐2H3 cell degranulation model, and a passive cutaneous anaphylaxis (PCA) mice model, and the structure changes were detected by circular dichroism (CD) spectra and ultraviolet (UV) spectra. The results showed that the processed egg proteins were more readily digested compared to raw egg proteins. The serological experiments revealed a significant reduction in immunoglobulin E binding of egg proteins after thermal treatments (p < 0.05), particularly after frying. Subsequently, the RBL‐2H3 cell degranulation experiment demonstrated a marked decrease in the level of egg allergens‐induced β‐hexosaminidase release after cooking (p < 0.05). Moreover, the results from the PCA mice model indicated that the increase in vascular permeability was effectively relieved in the treated groups, especially in frying group (p < 0.05). Additionally, the α‐helix and β‐turn contents of processed egg proteins were significantly decreased (p < 0.05) compared with native egg proteins. The UV spectra findings showed that all cooking treatments caused significant alterations in the tertiary structure, and fluorescence analysis indicated that cooking decreased the surface hydrophobicity of egg proteins. In conclusion, four traditional cooking methods reduced the allergenicity of egg proteins, particularly frying, and this reduction was associated with structural changes that could contribute to the destruction or masking of epitopes of egg allergens. Practical Application Egg allergy has a serious impact on public health, and there is no ideal treatment method at present. This study demonstrated that four traditional cooking methods (boiling, steaming, baking, and frying) reduced the allergenicity of egg proteins, especially frying, and the results will provide a basis for the development of hypoallergenic egg products.
... Conversely, hydrolyzed rice protein, another plant protein source permitted in IF, appears to be a safer option for infants with CMA (Vandenplas, Brough, et al., 2021). Besides rice being relatively hypoallergenic, an additional reason is the enzymatic hydrolysis that reduces the allergenicity of intact proteins (Pali-Schöll, Untersmayr, Klems, & Jensen-Jarolim, 2018). Research indicates that 92 infants with IgE-mediated CMA were well tolerating rice protein-hydrolyzed IF (Reche et al., 2010). ...
... Найчастішим шляхом сенсибілізації є травна система, коли рибні антигени швидко всмоктуються, тому порушення денатураційної функції шлункового соку може призводити до часткового перетравлення і підвищення рівня антигенних пептидів [24]. У працівників рибнопереробних підприємств алергія проявляється в разі контакту з рибою у вигляді кропив'янки, екземи або анафілаксії [30,40]. Існує зв'язок між порушенням бар'єрної функції шкіри через зниження синтезу філагрину в дітей з атопічним дерматитом (АД) та ризиком розвитку ХА на рибу та морепродукти [3,23]. ...
Article
Full-text available
In recent years, about 4% of children and 1% of adults in the world have suffered from food allergies to fish and seafood. About 40% of children have a high level of threat of systemic reactions. Literature data on the prevalence of FA on fish and seafood in the world are summarized, the principles of modern diagnostics are defined, taking into account the possibilities of molecular component diagnostics of fish and seafood allergen molecules. Purpose – To analyze the prevalence of allergies to fish and seafood in children; determine the structure of sensitization to the main allergens of these products using molecular component diagnostics. Materials and methods. 29 children aged from 3 to 18 years with clinical signs of allergies to fish and seafood were examined, with confirmation of sensitization to them using the ALEX-2 molecular component diagnostic method (Macro Array Diagnostics GmbH, Austria). Levels of specific IgE were determined. Results. Among those examined, children with food allergies to fish and seafood aged 3-6 years predominated (41.4%). 18 (62.1%) children had a severe allergy history. 19 (65.5%) children had oral allergy syndrome; 18 (62.1%) children had a papular rash with itching. Difficulty breathing was observed in 16 (55.2%) children. About half of the subjects were sensitized to fish allergens, most often to β-parvalbumin. Only 4 (13.8%) children were sensitized to fish enolase and eldolase. Among seafood, sensitization to squid predominated in children - in 18 (62.1%) children, while sensitization to tropomyosin was low - 1 (3.4%) case. Conclusions. The problem of the appearance of fish and seafood is relevant today, especially among children aged 3-6 years (41.4%). In children with fish allergy, sensitization to β-parvalbumin of different fish species predominated, namely to salmon (51.7%), mackerel (51.7%), herring (48.3%), carp (48.3%), tuna (48.3%), cod (41.4%). As a result of sensitization to seafood, sIGE was more often recorded to extracts of squid (62.1%), oyster (27.6%), lobster (20.7%), mussels (20.7%), clam (17.2%). The research was carried out in accordance with the principles of the Helsinki Declaration. The study protocol was approved by the Local Ethics Committee of the participating institution. The informed consent of the patient was obtained for conducting the studies. No conflict of interests was declared by the authors.
... For 12 allergenicity assessment the most reliable tests involve oral food challenge, (Costa, et al., 2022). Moreover, a good parameter for the characterization of allergens is their stability under gastric conditions (Moreno, 2007;Pali-Schöll, Untersmayr, Klems, & Jensen-Jarolim, 2018), and in vitro tests of pepsin digestion was incorporated since 2001 into a FAO/WHO procedure for the allergenicity assessment of novel food proteins (FAO, 2001). Enzymatic protein hydrolysates are a good alternative to intact proteins for obtaining special formulations to provide nutritive support to specific population groups such as elderly, infants, and food-allergic patients. ...
Preprint
Full-text available
Hazelnut, pistachio and cashew are tree nuts with health benefits but also with allergenic properties being prevalent food allergens in Europe. The allergic characteristics of these tree nuts after processing combining heat, pressure and enzymatic digestion were analyzed through in vitro (Western blot and ELISA) and in vivo test (Prick‐Prick). In the analyzed population, the patients sensitized to Cor a 8 (nsLTP) were predominant over those sensitized against hazelnut seed storage proteins (Sprot, Cor a 9 and 14), which displayed higher IgE reactivity. The protease E5 effectively hydrolyzed proteins from hazelnut and pistachio, while E7 was efficient for cashew protein hydrolysis. When combined with pressured heating (autoclave and Controlled Instantaneous Depressurization (DIC)), these proteases notably reduced the allergenic reactivity. The combination of DIC treatment before enzymatic digestion resulted in the most effective methodology to drastically reduce or indeed eliminate the allergenic capacity of tree nuts.
Article
Introduction: Acid suppression medications, such as proton-pump inhibitors (PPIs) and histamine-2 receptor antagonists, are commonly prescribed for the treatment of gastroesophageal reflux disease and other gastrointestinal disorders. However, concerns regarding potential long-term side effects are brought up by the overuse of PPIs. This study aimed to investigate the relationship between PPI usage, allergy, and asthma in the general US population. Methods: Data of individuals aged ≥20 years who had information on PPI use and questionnaires on allergy and asthma in the US National Health and Nutrition Examination Survey (NHANES) 2005-2006 were analyzed. Univariate and multivariable logistic regression analyses were performed to determine the associations between PPI use, prevalent allergy, and asthma. Results: A total of 4,481 participants (representing 198,543,007 US individuals after weighting) were included in the analyses. PPI use was not significantly associated with the presence of allergy or asthma in the general study population after adjustment. However, in females without steroid exposure, PPI use was significantly associated with increased odds of allergy (adjusted odds ratio [aOR] = 1.69, 95% confidence interval [CI]: 1.002-2.86), among which esomeprazole use was significantly associated with increased odds of allergy (aOR = 2.68, 95% CI: 1.30-5.54) and lansoprazole with increased odds of asthma (aOR = 3.44, 95% CI: 1.50-7.87) as compared to no PPI use. Duration of PPI use was not significantly associated with allergy or asthma. Conclusions: In US women without steroid exposure, PPI use is associated with increased likelihood of prevalent allergy and asthma.
Article
Full-text available
BACKGROUND: Early introduction of dietary peanut in high-risk infants with severe eczema, egg allergy, or both prevented peanut allergy at 5 years of age in the Learning Early About Peanut Allergy (LEAP) study. The protective effect persisted after 12 months of avoiding peanuts in the 12-month extension of the LEAP study (LEAP-On). It is unclear whether this benefit is allergen and allergic disease specific. Objective: We sought to assess the effect of early introduction of peanut on the development of allergic disease, food sensitization, and aeroallergen sensitization. METHODS: Asthma, eczema, and rhinoconjunctivitis were diagnosed based on clinical assessment. Reported allergic reactions and consumption of tree nuts and sesame were recorded by questionnaire. Sensitization to food allergens and aeroallergens was determined by means of skin prick testing and specific IgE measurement. RESULTS: A high and increasing burden of food allergen and aeroallergen sensitization and allergic disease was noted across study time points; 76% of LEAP participants had at least 1 allergic disease at 60 months of age. There were no differences in allergic disease between LEAP groups. There were small differences in sensitization and reported allergic reactions for select tree nuts, with levels being higher in the LEAP consumption group. Significant resolution of eczema and sensitization to egg and milk occurred in LEAP participants and was not affected by peanut consumption. CONCLUSION: Early consumption of peanut in infants at high risk of peanut allergy is allergen specific and does not prevent the development of other allergic disease, sensitization to other food allergens and aeroallergens, or reported allergic reactions to tree nuts and sesame. Furthermore, peanut consumption does not hasten the resolution of eczema or egg allergy.
Article
Full-text available
Objective: To highlight challenges associated with this novel preventive strategy. Data sources: The Learning Early About Peanuts (LEAP) and Enquiring About Tolerance (EAT) Studies, with reference to other oral tolerance induction studies. Study selections: Randomized clinial trials seeking to prevent food allergy through allergen introduction in infancy. Results: Oral tolerance induction programs that use a regimen of consumption of 2 g/week of protein are effective in preventing peanut and egg allergy. LEAP findings suggest oral tolerance induction is allergen specific. Adding peanut and other common food allergens (egg, fish, sesame, milk) to the infant diet has no adverse nutritional or growth effects and does not increase rates of food allergy. Breastfeeding rates are not adversely affected by these interventions. In the Western world, nonwhite children have the highest risk of food allergy, but their families are the least likely to participate in oral tolerance induction programs. Conclusion: Many challenges must be overcome to implement successful food allergy prevention strategies. Allergy testing of high-risk infants (those with moderate to severe eczema and/or egg allergy) before commencing oral tolerance induction is desirable, but access is not universal. Dietary interventions would ideally be implemented in infancy before allergic sensitization and allergy occur, using a program that provides protection against multiple common allergens. Further research and consensus with regard to food preparations, target populations, dosing regimens, and preparations and clearly defined adherence are now required.
Article
Full-text available
Background: With obesity continuing as a global epidemic and therapeutic technologies advancing, several novel endoscopic and minimally invasive interventions will likely become available as treatment options. With improved technologies and different treatment strategies, as well as different patient populations being targeted, there will be greater application in the treatment armamentarium of specialists dedicated to treating obesity. We sought to review the existing technology and provide a review. Methods: Literature review was carried out for endoscopic and minimally invasive devices. Some of these products are not FDA approved, so limited data are available in their review. Results: A summary of the device and data currently available on weight loss and safety profile is provided. Several products are in clinical trials or will be soon. Some of the technology has limited data and companies will be submitting their results for FDA evaluation. Conclusions: The obesity epidemic and associated weight-related diseases represent a tremendous burden to health care practitioners. As such, a multi-modal and progressive approach, with data and outcomes examined, is likely the best and most comprehensive method to care for these patients. SAGES endorses the benefits of minimally invasive and endoscopic approaches in the treatment of obesity and its related co-morbidities.
Article
Full-text available
Starch is a major source of energy in the human diet and is consumed in diverse forms. Resistant starch (RS) escapes small intestinal digestion and is fermented in the colon by the resident microbiota, with beneficial impacts on colonic function and host health, but the impacts of the micro- and nanoscale structure of different physical forms of food starch on the broader microbial community have not been described previously. Here, we use a porcine in vitro fermentation model to establish that starch structure dramatically impacts microbiome composition, including the key amylolytic species, and markedly alters both digestion kinetics and fermentation outcomes. We show that three characteristic food forms of starch that survive digestion in the small intestine each give rise to substantial and distinct changes in the microbiome and in fermentation products. Our results highlight the complexity of starch fermentation processes and indicate that not all forms of RS in foods are degraded or fermented in the same way. This work points the way for the design of RS with tailored degradation by defined microbial communities, informed by an understanding of how substrate structure influences the gut microbiome, to improve nutritive value and/or health benefits. IMPORTANCE Dietary starch is a major component in the human diet. A proportion of the starch in our diet escapes digestion in the small intestine and is fermented in the colon. In this study, we use a model of the colon, seeded with porcine feces, in which we investigate the fermentation of a variety of starches with structures typical of those found in foods. We show that the microbial community changes over time in our model colon are highly dependent on the structure of the substrate and how accessible the starch is to colonic microbes. These findings have important implications for how we classify starches reaching the colon and for the design of foods with improved nutritional properties.
Article
Full-text available
Significance Our results show that a standardized laboratory psychosocial stressor causes a greater inflammatory response in young healthy participants with an urban upbringing in the absence of pets, relative to young healthy participants with a rural upbringing in the presence of farm animals. In view of the known links between persistent inflammatory states and psychiatric disturbances, and considering that many stress-associated physical and mental disorders are more prevalent in environments offering a narrow range of microbial exposures, we feel that our findings are of general interest and significance. Moreover, we feel our study is timely, as urbanization and the associated socioeconomic consequences are increasing.
Article
Roasting has been implicated in the increase of peanut allergenicity due to the chemical reactions that occur during the process. However, this increase is not fully understood, and little information is available regarding the role of roasted peanut allergens in the initial phase of allergy, where dendritic cells (DCs) play a key role. We sought to analyze differences in the internalization of Ara h 3 from raw and roasted peanut by immature monocyte-derived DCs (MDDCs) and the implication of the mannose receptor in the uptake. Ara h 3 was purified from raw and roasted peanut (Ara h 3-raw and Ara h 3-roas) and labeled with a fluorescent dye. The labeled allergens were added to MDDCs obtained from 7 donors and internalization was analyzed after 10, 30, and 120 min by flow cytometry. In parallel, mannan, which blocks the mannose receptor, was added 30 min before adding the labeled allergens. Results showed that the internalization of Ara h 3-roas by MDDCs was significantly increased at every time point. However, the increase in the internalization of Ara h 3-raw was only significant after 2 h of incubation. Ara h 3-roas had an enhanced capacity to be internalized by MDDCs in comparison with Ara h 3-raw at every time point. Blocking the mannose receptor decreased the internalization of Ara h 3-roas but not Ara h 3-raw. In conclusion, the internalization of Ara h 3-roas by the MDDCs is enhanced when compared to Ara h 3-raw, and the mannose receptor might be implicated in this enhancement.
Article
Starch is the major source of dietary glucose for rapid development of children. Starches from various crops naturally differ in molecular structures and properties. Cooking, processing, and storage may change their molecular properties and affect their digestibility and functionality. Starch digestion is affected by its susceptibility to α-amylase and α-glucosidase (maltase), and the susceptibility is determined by starch granule architecture and glucan structures, as well as the interaction between starch and other food components. Starch is given as a complementary feeding to young children in many cultures, and starch or modified starch, is used in special formulae of infant foods or supplements. Although indigestible starch does not provide much energy, it can benefit colonic health.
Article
Based on the developmental physiology of pancreatic amylase production, starch digestion in young infants was anticipated to be compromised whenever compared with that in older infants and toddlers. This appears to be the case, but with great variability among infants to digest starch. Evidence points to the importance of maltase-glucoamylase in young infants and its effect on starch digestion. These observations have critical importance for recommendations regarding the feeding of starch-containing foods to young infants.
Article
Until now there is no molecular model of starch digestion and absorption of the resulting glucose molecules along the larval midgut of Musca domestica. For addressing to this, we used RNA-seq analyses from seven sections of the midgut and carcass to evaluate the expression level of the genes coding for amylases, maltases and sugar transporters (SP). An amylase related protein (Amyrel) and two amylase sequences, one soluble and one with a predicted GPI-anchor, were identified. Three highly expressed maltase genes were correlated with biochemically characterized maltases: one soluble, other glycocalyx-associated, and another membrane-bound. SPs were checked as being apical or basal by proteomics of microvillar preparations and those up-regulated by starch were identified by real time PCR. From the 9 SP sequences with high expression in midgut, two are putative sugar sensors (MdSP4 and MdSP5), one is probably a trehalose transporter (MdSP8), whereas MdSP1-3, MdSP6, and MdSP9 are supposed to transport glucose into cells, and MdSP7 from cells to hemolymph. MdSP1, MdSP7, and MdSP9 are up-regulated by starch. Based on the data, starch is at first digested by amylase and maltases at anterior midgut, with the resulting glucose units absorbed at middle midgut. At this region, low pH, lysozyme, and cathepsin D open the ingested bacteria and fungi cells, freeing sugars and glycogen. This and the remaining dietary starch are digested by amylase and maltases at the end of middle midgut and up to the middle part of the posterior midgut, with resulting sugars being absorbed along the posterior midgut.
Article
A growing concern around the world is the number of people who are suffering from food protein allergies. One potential approach to decrease protein allergenicity is to block IgE-binding epitopes of the protein allergen by attachment of polysaccharides via the Maillard reaction (i.e., glycation). Protein glycation has been extensively studied to modify various functional properties. We wanted to examine whether glycates could reduce IgE binding in patients with cow milk protein allergy and to explore how the size (molar mass; MW) of the polysaccharide affects this IgE-binding capacity. Glycation was performed using the initial step of the Maillard reaction performed in aqueous solutions. The specific goal of this study was to reduce the IgE-binding capacity of whey protein isolate (WPI) through glycation with dextran (DX). Blood sera were obtained from 8 patients who had been diagnosed with cow milk protein allergy, and a composite sera sample was used for IgE-binding analysis by the ImmunoCap (Phadia, Uppsala, Sweden) method. The WPI was glycated with DX of MW ranging from 1 to 2,000 kDa, and the MW of purified glycates was determined using size-exclusion chromatography coupled with multiangle laser light scattering. The WPI to DX molar ratios in the glycates made from DX that had MW values of 1, 3.5, 10 (G10), 150, 500, and 2,000 kDa were 1:4, 1:3, 1:2, 1:1.5, 1:1, and 1:1, respectively. With the increase in the MW of DX, there was an increase in the MW values of the corresponding glycates but a decrease in the number of bound DX. The WPI-DX glycates had lower whey protein IgE-binding capacity than native WPI, with the lowest IgE-binding capacity obtained in the G10 glycate. The DX binding ratios and morphology results from atomic force microscopy images suggested that glycation of WPI with small-MW DX resulted in extensive protein surface coverage, probably due to the attachment of up to 4 DX molecules per whey protein. The lower IgE binding of the G10 glycate was likely due to greater steric hindrance (or a physical barrier) at the surface of the protein. In summary, our results demonstrate that glycating WPI with DX via Maillard reaction can potentially be used to decrease the allergenicity of whey protein.