You are on page 1of 11

Plant Foods for Human Nutrition

https://doi.org/10.1007/s11130-020-00800-8

REVIEW ARTICLE

Oats as a Safe Alternative to Triticeae Cereals for People


Suffering from Celiac Disease? A Review
Klára Kosová 1 & Leona Leišová-Svobodová 1 & Václav Dvořáček 1

# Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
Oats represent a promising alternative to small-grain cereals from Triticeae group (wheat, barley, rye) for persons suffering from
any form of gluten intolerance, especially celiac disease (CD), since oat-specific prolamins avenins reveal generally lower gluten
content and immunoreactivity. Recent studies on avenin molecular structure revealed large genetic variability in avenin se-
quences affecting the spectrum of gluten peptides produced by hydrolases in human digestive tract. The aim of the present
review is to summarise recent knowledge obtained in laboratory studies focused on the effect of avenin-derived peptides on
reactivity of crucial components of human immune system such as dendritic cells (DC) and T-cells. The other part of the review
summarises the results of clinical studies with CD patients including oat products in their diet. Since different clinical studies
revealed contradictory results regarding potential safety of oats for CD patients, the focus has to be directed at genetic variability
in oat avenins. Identification of avenin isoforms with minimum CD immunoreactivity will open up ways leading to designing
novel oat cultivars suitable for CD patients. Knowledge on immunoreactivity of gluten peptides together with breeding new oat
cultivars revealing minimum avenin immunoreactivity with respect to CD as well as application of food processing technologies
leading to gluten content reduction should result in development of gluten-free oats safe for celiacs.

Keywords Celiac disease . Avenins . Genetic variability . Immunoreactivity . Clinical studies . Oats (Avena sativa)

Abbreviations SCFA short chain fatty acid


AA amino acid tTG2 tissue transglutaminase 2
CD celiac disease γIFN interferon γ
DC dendritic cells
E glutamic acid
GFD gluten-free diet
GI gastrointestinal
Introduction
Glt glutenin
Celiac disease (CD) is an autoimmune enteropathy triggered
IL interleukin
by dietary gluten in genetically predisposed persons carrying
LMW low-molecular weight
HLA-DQ2.2/2.5/8 receptors on the membranes of dendritic
moAb monoclonal antibody
cells (DC). Gluten is a mixture of hardly-hydrolysable poly-
P proline
peptides rich in proline and glutamine which arises by hydro-
Q glutamine
lysis of grain storage proteins belonging to prolamin family in
wheat (glutenins and gliadins), barley (hordeins), rye
Electronic supplementary material The online version of this article
(https://doi.org/10.1007/s11130-020-00800-8) contains supplementary
(secalins), and oats (avenins) [1]. Grain storage proteins in-
material, which is available to authorized users. cluding albumins, globulins, prolamins, glutelins, and
unextractable protein fractions are estimated to amount ap-
* Klára Kosová proximately 15% of total kernel weight in small-grain cereals
kosova@vurv.cz (wheat, barley, rye) out of which prolamins are estimated to
contribute roughly 15% of total grain storage protein weight,
1
Division of Crop Genetics and Breeding, Crop Research Institute, i.e., approximately 6–8% of total kernel weight. The name
Prague6-Ruzyně, Czechia prolamin is derived from proline and amine/amide referring
Plant Foods Hum Nutr

to amide groups in glutamine residues. The joint proline (P) autoantibodies, γIFN, interleukins IL-15, IL-21. Histological
and glutamine (Q) content can reach up to 50–60% of total symptoms of CD include damage to lamina propria (mucosa)
amino acids in wheat gliadins [2]. Prolamins differ in their in duodenum leading to villous atrophy [1, 7, 8].
solubility in ethanol and electrophoretic mobility as well as It is estimated that the amount of people sufferring from
their ability to form oligomers which is determined by the gluten intolerance can reach up to 1% in Western countries
number of cysteine residues enabling formation of intra- population. The only live-long efficient treatment lies in con-
and inter- molecular disulphide bonds. In wheat, prolamins summation of gluten-free diet (GFD) which is a serious prob-
are distinguished into alcohol-insoluble oligomeric lem in Western countries due to ubiqity of gluten as an addi-
glutenin fraction [high and low molecular weight (LMW) tive in food industry. Contamination of gluten-free foods with
glutenins] and alcohol-soluble monomeric gliadin fraction gluten represents another important problem. Oats can repre-
containing S-rich α-, β, and γ-gliadins, and S-poor ω-gli- sent a promising alternative to foods prepared from wheat
adins. S-rich prolamins contain three conserved regions A, (barley, rye) since the majority of celiacs can tolerate oats
B, and C interspersed by repetitive regions and variant without any clinical symptoms [4]. Currently, oats is included
regions while S-poor prolamins lack the conserved re- into gluten-free ingredients, i.e., those ingredients which do
gions. S-poor prolamins such as wheat ω-gliadins and bar- not exceed 20 mg gluten per 1 kg of food (20 ppm), by
ley C hordeins were suggested to have evolved from S-rich European Commission Regulation 41/2009 [9] although its
ones by deletion of the conserved regions and multiplica- safety for celiacs is still a matter of debate. In Canada, con-
tion of repetitive regions [1]. sumption of pure and uncontaminated oats where gluten con-
Celiac disease represents a complex of human body re- tent does not exceed 20 mg/kg as detected by R5-ELISA test
sponses to gluten intolerance which is underlied by the inabil- was recommended by the Canadian Celiac Association,
ity of human small intestine enzymes to cleave peptide bonds Canadian Food Inspection Agency, and Health Canada [10].
in P + Q rich sequences [1, 2]. CD is determined genetically In contrast, oat is not considered „gluten-free food “in
by the presence of HLA-DQ2.2, HLA-DQ2.5 or HLA-DQ8 Australia and New Zealand [11]. Recent studies have indicat-
haplotypes of receptors on the plasma membrane of dendritic ed significant genetic variability within cultivated oats with
cells (DCs) which are important antigen-presenting cells [3]. respect to oat avenin molecular structure, potential immuno-
A crucial role in CD biogenesis plays human enzyme tissue reactivity and induction of CD-related clinical symptoms
transglutaminase 2 (tTG2) which deamidates glutamine (Q) to (reviewed in [12, 13]).
glutamic acid (E) thus introducing a negative charge into QXP The aim of the present review was to summarise recent
motifs in gluten peptides and enhancing their interaction with knowledge on oats avenins molecular structure, genetic vari-
HLA-DQ2/8 receptors. The interaction of HLA-DQ2/8 with ability and potential immunoreactivity with respect to CD.
deamidated gluten peptide leads to formation of HLA-DQ2/8- Results of clinical studies aimed at determining the effects of
gluten complex which interacts with T cell receptor (TCR) on oats on key steps in induction of CD symptoms are
the surface of specific CD4+ T lymphocytes in lamina propria summarised. Currently, it is necessary to explore oat genetic
of proximal small intestine. The interaction leads to T cell variability with respect to avenin molecular structure and their
proliferation and release of γ-interferon (γIFN) and interleu- potential immunoreactivity. Recommendations for oat
kin 21 (IL-21) leading to inflammatory processes resulting in breeders with respect to desired avenin structure inducing no
villous atrophy [1, 4]. Moreover, CD is also an autoimmune (minimum) immunoractivity should be formulated. The
disease due to tTG2 transamidation activity leading to a for- knowledge on the relationship between avenin molecular
mation of complexes between tTG2 and gliadin peptides structure and their potential immunoreactivity opens up new
which can bind to HLA-DQ2.2/2.5. and induce specific ways for genetic modifications of avenin sequences.
IgA-class autoantibodies against tTG2. [1, 4]. The strongest
immune response was found for 33-mer peptide (p57–89)
derived from wheat α2-gliadin repetitive region. Currently, Avenin Molecular Structure and Phylogenetic
six different epitopes recognised by HLA-DQ2.5 receptors Analysis
were identified in α2-gliadin 33-mer [1, 5]. Besides the 33-
mer (p57–89), the 25-mer (p31–43) interacting with interleu- Oats (Avena sativa L.) belongs to gramineaceous group called
kin 15 (IL-15) was identified in α2-gliadin [1]. Only three Avenae which is related but distinct from Triticeae group in-
epitopes recognised by HLA-DQ8 were identified; one in α- cluding wheat, barley, and rye. The genus Avena includes up
gliadins, another one in γ-gliadins, and the third one in to 30 species with diploid, tetraploid and hexaploid genomes;
glutenins [3]. Moreover, HLA-DQ8-mediated CD seems to cultivated oats (Avena sativa) is a hexaploid species with
be independent on tTG2 mediated deamidation [6]. AACCDD genomes. Oat grain represents a valuable source
Molecular markers of CD include serum antibodies against of a wide array of nutrients including β-glucans as an impor-
gluten-derived peptides (IgE, IgG); tTG2-targeted IgA class tant source of fiber, lipids, avenanthramides, and grain storage
Plant Foods Hum Nutr

proteins including globulins, albumins, prolamins, and with large P + Q rich repetitive regions. Avenin sequences
glutelins [4, 14, 15]. Oat prolamins are called avenins. In belonging to groups B and C contain eight cysteine residues
comparison to Triticeae prolamins reaching 6–8% of kernel while avenins of group A have nine cysteine residues [2].
weight, avenins represent only about 1.2–2% of kernel weight However, in comparison to wheat prolamins, avenins can
[2, 16]. Avenins also represent a lower fraction (around 13%) form only intermolecular disulfide bonds [1]. Avenins re-
of total oat grain proteins with respect to Triticeae prolamins vealed a lower joint P + Q content of roughly 30% of total
(up to 35% of total grain protein in wheat) since the major oat amino acid content than compared to wheat α- and γ-gliadins
grain storage proteins are globulins [4, 17]. whose P + Q joint content can reach up to 50–60% of total
amino acid content [1, 2, 22]. Thus, it can be expected that
Avenin Gene Number Currently, oat genome sequencing pro- avenins are more easily hydrolyzable by duodenal enzymes.
ject is run by people from Aberystwyth University (https:// Moreover, it is becoming evident that oat genotypes differ in
avenagenome.org) using the diploid donors of the individual their avenin composition and that some oat genotypes could
genomes: A genome from Avena atlantica, C genome from lack some avenin structural types. In their study aimed at
Avena eriantha, and a putative D genome. However, a avenin analysis in selected Spanish oat lines, Real et al. [2]
complete oat genome sequence has yet to be published, thus identified avenins belonging to all A, B and C classes in all
the final number of avenin genes remains unknown. Chesnut lines except for OP722 which lacks class A avenins.
et al. [18] estimated around 25 avenin genes encoded by A. Expression pattern of avenin proteins during oat kernel
sativa genome while Londono et al. [19] detected only 10 and development was also studied. Chesnut et al. [18] detected
11 avenin genes expressed in A. sativa cvs Gigant and Dancer, avenin mRNAs and proteins between 4 and 6 days after an-
respectively. Currently, 54 avenin protein sequences (partial or thesis (DPA) with a maximum at 8 DPA while Real et al. [2]
full-length sequences) from Avena sativa and 19 avenin se- were able to detect avenin proteins at 8 DPA with a maximum
quences from other Avena species are available in NCBI da- expression level between 20 to 28 DPA. Greater expression
tabase (www.ncbi.nlm.nih.gov; downloaded on 30th levels were found for α- and γ-gliadin-like avenins of groups
September, 2019). B and C than for low-molecular-weight glutenins-like avenins
of group A.
Avenin Molecular Structure Similarly to Triticeae prolamins,
avenin molecules contain a signal peptide at N-terminal part
followed by the first repetitive region followed by conserved Avenin CD Immunoreactivity
A and B regions followed by second repetitive region and C
conserved region near to C-terminal part of the molecule. P + Avenin CD Reactive Epitopes and their Crossreactivity
Q rich repetitive regions can be deamidated by tissue with Anti-Gluten Antibodies
transglutaminase 2 (tTG2) to glutamic acid (E) to form immu-
noreactive epitopes in some types of avenin molecules. In Sollid et al. [3] provided nomenclature of gluten epitopes
contrast to S-poor gliadins which have only one long repeti- which react with the individual HLA-DQ receptors including
tive region, avenins have two shorter repetitive regions thus HLA-DQ2.2, HLA-DQ2.5, HLA-DQ8 and HLA-DQ8.5 re-
their hydrolysis yields shorter P + Q rich gluten-like peptides ceptor types. Two avenin-specific 9-mer epitopes named
[1, 2, 19]. DQ2.5-ave-1a (formerly known as Av-α9A) PYPEQQEPF
and DQ2.5-ave-1b (formerly known as Av- α9B)
Phylogenetic Analyses of Avenin Proteins Cluster analysis PYPEQEQPF located in the first P + Q rich repetitive region
based on avenin protein sequences revealed three groups were identified as HLA-DQ2.5 immunoreactive epitopes in
termed avenins A, B, and C, respectively (Fig. 1; [2]). some groups of avenin proteins based on the results of previ-
Cluster corresponding to group C contains two sub-clusters ous studies [7, 23]. In addition, 9-mer PFVQQQQPF se-
with high level of bootstrap values (Fig. 1). They differ by the quence formerly known as Av-γ9B epitope is located down-
presence of avenin-specific T cell epitopes in the first con- stream of DQ2.5-ave-1a in group III avenins [19].
served region [19]. Avenins B and C are homologous to wheat Recently, it is becoming evident that avenin molecules
alcohol-soluble S-rich α- and γ-gliadins, barley B hordeins reveal differences in their CD reactivity determined by
and rye γ-secalins, respectively, while avenins A are various kinds of assays including immunoreactivity assays
alcohol-insoluble proteins homologous to wheat LMW based on crossreactivity of some avenin sequences with
glutenins which means that they form oligomers due to disul- primary antibodies raised against wheat gluten epitopes,
fide bridges between cysteine residues. However, A-class assays based on avenin-induced agglutination of K562
avenins can be solubilised by using reduction agents indicat- cells, as well as avenin potential stimulatory effects on
ing that their alcohol insolubility is underlied by intermolecu- dendritic cells (DC), T cells, and other components of hu-
lar disulfide bonds. Oats does not contain S-poor prolamins man immune system.
Plant Foods Hum Nutr

Fig. 1 Molecular phylogenetic


analysis computed by Maximum
likelihood method based on the
Jones-Taylor-Thornton (JTT)
matrix-based model selected ac-
cording to AIC criterion [20]. The
tree with the highest log likeli-
hood (−1249.26) is shown. The
percentage of trees in which the
associated taxa clustered together
is shown next to the branches.
Initial tree(s) for the heuristic
search were obtained automati-
cally by applying Neighbor-Join
and BioNJ algorithms to a matrix
of pairwise distances estimated
using a JTT model, and then
selecting the topology with supe-
rior log likelihood value. A dis-
crete gamma distribution was
used to model evolutionary rate
differences among sites (5 cate-
gories (+G, parameter = 0.6862)).
The rate variation model allowed
for some sites to be evolutionarily
invariable ([+I], 0.00% sites).
Evolutionary analyses were con-
ducted in MEGA7 [21]

Immunoreactivity Assays Londono et al. [19] proposed a crossreactivity of wheat


G12 and R5 moAb with avenins since four R5-related epi-
Antibodies Used for Testing Anti-Gluten Immunoreactivity tope variants and six G12-related epitope variants are pres-
ent in sequences present in avenin-specific epitopes
Antibodies used for gluten peptides detection in oats in- DQ2.5-ave-1a (formerly known as Av-α9A) and DQ2.5-
clude the two major kinds of anti-gluten monoclonal an- ave-1b (formerly known as Av-α9B), respectively. No per-
tibodies R5 moAb and G12 moAb, respectively. The R5 fect gluten epitopes were found in avenins; however, se-
moAb antibody binds to epitope variants QQPFP, quences revealing a substitution of one or two amino acids
QQQFP, LQPFP, and QLPFP, and, to a lesser extent, to were recognised by the antibodies raised against wheat gluten
QLPTF, QQSFP, QQTFP, PQPFP, and QQPYP, and is peptides. Regarding wheat R5 responsive epitopes, avenin
mostly used for detection of gluten in wheat, barley and epitopes QQPFL, QQPFV and QQPFM revealing a
rye [24]. Sandwich R5 ELISA assay was developed for a crossreactivity with wheat epitope QQPFP and avenin se-
rapid screening of gluten [25, 26]. quence YQPFYP revealing a crossreactivity with wheat epi-
Another kind of anti-gluten antibody represents G12 moAb tope QQPYP were identified. Regarding wheat G12 respon-
which binds to epitopes QPQLPY, QPQQPY, and QPQLPF sive epitopes, avenin sequence QPQLQ revealing a
present in 33-mer (57–89) derived from α2 gliadin [27]. G12 crossreactivity to wheat QPQLPY, avenin sequences
mAb reveals limited cross-reactivity with avenins. An ELISA QPQQQA, QQQQPF, QPQQQA, QQQQPF, QPQQLP and
assay using G12 moAb was developed for a rapid screening of QPQLPF revealing a crossreactivity to wheat QPQQPY, and
gluten [28]. avenin sequence QPQLQL revealing a crossreactivity to
Plant Foods Hum Nutr

wheat QPQLPF were identified by Londono et al. [19] in immunoreactivity in avenins. Similar differences were also
avenin protein sequences from cvs Dancer and Gigant. found in immunoreactivity of avenin extracts using a spe-
Londono et al. [19] also reported the presence of one of the cific moAb against α20-gliadin epitope [33]. The obtained
two HLA-DQ2.5 responsive epitopes in 7 out of 11 avenin results led the authors to conclusion that oat breeding pro-
genes in cv Dancer and in 4 out of 10 avenins in cv Gigant. grams aimed at selection of CD-safe oat varieties with a
Londono et al. [19] also performed an analysis of avenin minimum immunoreactivity may be a realistic goal; how-
proteins in 13 Avena species including 12 diploid and tetra- ever, they also noted that true safety can be considered
ploid Avena species and a hexaploid Avena cultivar Gigant only by the results of clinical trials.
with respect to gluten epitopes. A total of 717 avenin protein Ballabio et al. [34] found out differential response of 36 oat
sequences representing 78 different genes were obtained from cultivars to anti-gliadin protein kit using RIDASCREEN
the 13 Avena species indicating that the individual oat species ELISA test based on R5 moAb. The majority of studied oat
encode 7 to 10 avenin genes; the following sequence analysis cultivars revealed levels of gluten-like proteins well below the
revealed four avenin phylogenetic groups. Avenin sequence limit of 20 mg/kg although some varieties such as Expander,
analysis with respect to CD immunoreactivity revealed the HJA76037N, Kynon and BG2 revealed concentration of
presence of two avenin-specific T cell recognized epitopes gluten-like proteins very close to 20 mg/kg, and a few oat
which were present once only per protein molecules and were varieties such as commercial husked cv. Dakar, experimental
located in the first repetitive region; the epitope DQ2.5-ave-1b naked genotype BG3, naked cultivar Nave and genotype BG4
PYPEQQQPF was present in all avenin genes from group II significantly exceeded the 20 mg/kg limit for gluten-free
whereas the epitope DQ2.5-ave-1a PYPEQQEPF was present products.
in all genes from group III while avenin genes from groups I Similarly, Benoit et al. [35] tested 19 oat varieties given
and IV contained no T cell immunoreactive epitopes. their response to R5 moAb antibody assay; R5 epitope
However, the authors reported that none of the oat species LQPFP is present in highly immunogenic α-gliadin 33-mer
studied lacked group II or III avenins, i.e., they all possess at sequence. Most of the 19 oat cultivars tested revealed a neg-
least one of the two avenin-specific T cell epitopes. However, ative response to R5 assay; however, three of them including
it should be borne in mind that the genes present in a given Brazilian Check, Kanota, and Curt, revealed a positive re-
genome can be expressed at differential rates resulting in dif- sponse to R5 assay indicating presence of immunogenic pep-
ferent final protein levels; thus, the resulting levels of immu- tides. Moreover, the three oat cultivars also revealed a positive
nogenic proteins cannot be estimated from the presence of response to Skerritt antibody which revealed even higher glu-
genes or gene copies in the genome due to various mecha- ten levels when compared with R5 antibody (Supplementary
nisms regulating transcription and posttranscription processes. Table S1).
Later, Nassef et al. [29, 30] used electrochemical Recently, Giménez et al. [36] studied variability in avenin
immunosensors to detect gliadin peptides in raw and proc- CD immunoreactivity in a set of 95 oat varieties of
essed food using a monoclonal antibody against α-gliadin Mediterranean origin using individual avenin protein resolu-
56–75 region (CDC 5) or antigliadin Fab fragments (CDC5- tion by reversed-phase high performance liquid chromatogra-
Fab). Results obtained by this method correlate with ELISA phy (RP-HPLC) and testing avenin immunoreactivity by
determination and reveal high sensitivity, easy performance, using G12 moAb based ELISA tests. The researchers reported
and short assay time. a high variability in avenin peak patterns within the studied oat
In addition, moAb 401.21 also referred to as Skerritt anti- samples as well as differences in CD immunoreactivity among
body which binds to PQPQPFPQE and PQQPPFPEE epi- different avenin peaks. Overall, gluten levels determined by
topes can be used for gluten detection [31]. A brief overview G12-based ELISA did not exceed 20 mg/kg in the individual
of studies aimed at detection of immunoreactive peptides oat samples while only one sample exceeded 40 mg/kg.
varieties is given in Supplementary Table S1.
Mujico et al. [32] compared avenin immunoreactivity to Dendritic Cells (DC) Reactivity Assays
anti-LMW glutenin moAb recognizing PFVQ sequence
known as Glt-156 LMW epitope [23] which is found in Comino et al. [37] studied avenin proteins, their gluten pep-
two avenin epitopes QQPFVQQQPFVQQ and tides and their potential stimulatory effects on DC in a set of
QYQPYPEQQQPFVQ using immunoblot analysis of eth- oat cultivars from Spanish and Australian commercial re-
anol extracts and they found significant differences be- sources. They observed significant polymorphism patterns in
tween the oat cultivars tested. Wodan and Zwarte avenin fraction separated on 1D SDS-PAGE gels which was
President revealed the strongest reaction while Mansholt even greater than in globulin fraction. The large differences in
III revealed the faintest one which was in contrast with avenin protein sequences whose size ranged between less than
the results of T cell stimulatory assay. Thus, different as- 20 up to 50 kDa on 1D gels were also reflected at the level of
says can reveal different results regarding anti-gluten their hydrolysis products. With respect to wheat gliadins
Plant Foods Hum Nutr

producing long 33-mer P + Q rich peptide inducing immune events were studied: K562(S) cells agglutination,
response, oat avenins were found to yield a series of shorter transepithelial electrical resistance of T84-cell monolayers,
peptides (6, 10, 14, and 27 AA) which are easier to be cleaved intracellular levels of TG2 and phosphorylated form of protein
or internalised to endosomes in DCs than larger peptides from 42–44 in T84 cells. The tests revealed that Nave elicited these
wheat gliadins [37]. The short 6 AA avenin-derived gluten events whereas Irina and Potenza did not. These results cor-
peptides did not trigger any DC stimulatory effects. Only the related with the extent of pepsin-mediated proteolysis of the
27 AA avenin-derived peptides EF27 and QM27 triggered T prolamin fraction. In addition, the ability of a cultivar to in-
cell proliferative response similar to wheat gliadin-derived 33- duce the above-listed events correlated with avenin electro-
mer peptide. However, a different way of endosomal internal- phoretic pattern and their reactivity to anti-gliadin polyclonal
ization of shorter (6 AA) and intermediate (10 and 14 AA) antibodies.
peptides was proposed with respect to the larger ones. As a
result, these peptides do not induce T cell response leading to T Cell Reactivity Assays
development of CD molecular symptoms such as T cell pro-
liferation, γIFN release, tTG2 autoantibodies production, etc. Mujico et al. [32] found a variation in stimulatory capacity of
[37, 38]. specific T cell line from a celiac patient within a set of 26 oat
Immunoreactive peptides interacting with anti-33mer varieties of Dutch origin (Ascot, Astor, Charming, Charmoise,
monoclonal antibody moG12 raised against 33-mer gluten Dalguise, Dominik, Fervente, Firth, Freddy, Gambo, Gele van
peptide from wheat α2-gliadin as detected in oat avenins by Timmermans, Gerald, Gigant, Leanda, Mansholt III, Markant,
Comino et al. [37]: Mustang, Ouderwetse Zeeuwse Partij, Panache de Roye,
Immunoreactive peptides derived from gliadin-like avenins Powys, Sang, Troshaver uit Besel, Valiant, Wodan, Zandster,
(B- and C-type avenins): QL6 (QPQLQL), QQ6 (QPQLQQ), Zwarte President) compared to four synthetic peptides con-
P V 1 0 ( P Y P E Q Q E P F V ) , E F 2 7 taining two known avenin epitopes (QQPFVQQQQPFVQQ
(EQYQPYPEQQEPFVQQQPPFVQQEQPF). and QYQPYPEQQQPFVQ). The results of the T cell prolif-
Immunoreactive peptides derived from glutenin-like eration assay revealed that the majority of oat samples contain
avenins (A-type avenins): avenins possessing epitopes with T cell stimulatory capacity
QL14 (QQPFMQQQPFMQPL), QM27 but some samples (Dalguise, Gambo, Markant, Wodan,
(QYQPYPEQQPFMQQQQPFMQPLLQQQM). Zanster) revealed a minimum stimulatory capacity indicating
that they contain avenins with minimum gluten-like epitopes.
Agglutination Assays Arentz-Hansen et al. [7] studied T cell reactivity to synthet-
ic avenin-derived peptides and found recognition by two T
Silano et al. [39] compared avenins isolated from three oat cell lines for peptide 1490 with the following sequence
varieties given their ability to agglutinate K562 human mye- SEQYQPYPEQQEPFVQQQQ which was dependent on pep-
logenous leukemia cells and to disrupt rat liver lysosomes tide deamidation by tTG2. A 9-mer core peptide Av-α9A with
with respect to the activities of wheat gliadin and rice grain sequence PYPEQEEPF arising from deamidation of gluta-
proteins. A significant difference between the different oat mine to glutamic acid (E) at 6th position and revealing se-
varieties was found as avenins from Italian variety Astra and quence homology to wheat DQ2-α-I gliadin epitope
Australian variety Mortlock were much more active in K562 PFPQPELPY was predicted to be recognised as an epitope
cells agglutination and lysosome disruption than avenins iso- for HLA-DQ2 receptors on the surface of dendritic cells.
lated from Australian variety Lampton. Wheat gliadin (a pos- Comino et al. [38] found a direct relationship between
itive control) revealed enhanced activity with respect to all avenin reactivity to monoclonal AbG12 antibody raised
three avenins while rice protein (a negative control) did not against wheat α-2 gliadin-derived 33-mer (p57–89) and their
reveal measurable activity. Moreover, the same authors inves- potential immunotoxicity determined as peripheral blood
tigated the effect of avenins isolated from four oat cultivars mononuclear T cell proliferation and γIFN release.
Astra, Ava, Lampton and Nave on proliferation and γIFN Hardy et al. [16] found T cell reactivity to 20mers
release by peripheral lymphocytes isolated from ten celiac encompassing 9mer sequences closely related to avenin-
children. They found out that avenins from Ava and derived epitopes DQ2.5-ave-1a (PYPEQEEPF where E re-
Lampton exhibited a relatively strong immunogenic reaction sults from deamidation of Q39 in several native avenin se-
comparable to wheat gliadin while avenins from Astra and quences) and DQ2.5-ave-1b (PYPEQEQPF, deamidation of
Nave induced significantly lower but still detectable immuno- Q19 to E). Further immunogenic sequences include 9mer epi-
genic response [40]. topes derived from partially deamidated avenin peptide
Silano et al. [41] investigated the potential of different oat QYQPYPEQEQPILQQQ (deamidation of Q34 to E) that in-
varieties to elicit tTG2-derived events in some in vitro models cluded a 9mer amino-acid sequence differing from DQ2.5-
of CD. The following early gluten-induced TG2-dependent ave-1b by substitution of isoleucine for phenylalanine at
Plant Foods Hum Nutr

position 9. Moreover, they reported a crossreactivity between increases with their concentration. Recently, it was found
hordein-derived epitope DQ2.5-hor-3b and avenin epitopes out that human body immune response is enhanced with
DQ2.5-ave-1b and DQ2.5-ave-1c, respectively. Cross- an increasing concentration of gliadin 33-mer due to a
reactive T cells specific for DQ2.5-hor-3b and avenin epitopes formation of supramolecular oligomeric rod-like struc-
DQ2.5-ave-1b and DQ2.5-ave-1c are commonly mobilized in tures of polyproline II-type based on oligomerization of
HLA-DQ2.5+ CD patients when they eat barley, and also oats, type II β-sheets formed by monomeric 33-mers [42].
but less consistently and at lower frequencies than in barley. It However, breeding for CD-safe oat varieties with reduced
could be concluded that hordein-derived peptides functioned gluten content can bring several obstacles due to the fact that
as „priming “means, i.e., immunogenic response to avenin- avenin genes are distributed in the whole oat genome includ-
derived peptides was induced by hordein-derived peptides ing a vicinity of several resistance genes. For example,
since avenin peptides themselves had reduced binding stabil- Satheeskumar et al. [43] detected three avenin loci in the vi-
ity on HLA-DQ2.5 as compared to homologous hordein- cinity of Pc68 gene conferring resistance to Puccinia coronata
derived peptides. The researchers thus demostrated that food crown rust.
antigen cross-reactivity has significant immunological rele- Lerner [44] postulated the major problems associated with
vance in vivo which opens the possibility that other peptides oat consumption as a part of GFD including genetic variability
may induce pathogenic response in CD patients under some in avenin-derived gluten peptides among oat varieties as well
circumstances. as contamination of oat products by other gluten-containing
cereals resulting in exceeding the gluten limit of 20 mg/kg.
Summary to Avenin CD Reactivity Regarding the potential breeding of gluten-safe oat varie-
ties, it is necessary to describe the variability of avenin alleles
Contradictory results obtained by different clinical stud- with respect to the potential gluten peptides. In the next step, it
ies probably reflect genetic variability with respect to will be necessary to exclude potential unsuitable avenin alleles
gluten content and its immunoreactivity in oats used in from promising oat materials. Recently, a boom of gene
food industry. Variability in avenin composition includ- editing techniques including CRISPR/Cas, TALENs, and
ing variability in avenin-derived gluten peptides and their others (reviewed in [45]) could facilitate the gain of prospec-
immunoreactivity to anti-gliadin antibodies was found tive oat materials cumulating only suitable avenin alleles.
[37, 41]. Some recent studies revealed differential gluten
peptides with differential stimulatory capacity on DC Clinical Studies on the Effects of Oats on CD Clinical
which indicates that people with CD who want to in- Symptoms
clude oats in their diet should pay attention to oat genet-
ic resources used for food preparation [16]. It thus arises Clinical studies aimed at the effects of oat consumption
a challenge for oat breeders to select oat genetic mate- by persons sufferring from CD revealed contradictory
rials with no (minimum) CD reactivity without any neg- results with respect to the effect of oat-containing diet
ative effects on other oat qualities. Some celiac patients on CD symptoms. A detailed overview of clinical studies
can contain gluten-reactive or avenin-reactive intestinal T dealing with oats as a part of GFD in adults and children
cells without any clinical symptoms due to remission. is presented in the review of La Vieille et al. [46]. A
Oat genetic variability leads to a variability of avenin- wide array of CD-related symptoms including gastroin-
derived peptides which can potentially induce CD re- testinal symptoms, malabsorption, mucosal changes, se-
sponse. A series of HLA-DQ2.5 interacting avenin- rological responses and overall quality of life regarding
derived peptides were characterised; however, only longer people using oat as a part of GFD were reviewed by
peptides can induce inflammatory response similar to those Cohen et al. [4]. Basic data on the clinical studies
induced by wheat gliadin 33-mer, i.e., T-cells activation, discussed in the following text are provided in
production of serum IgA anti-avenin and anti-tTG2 anti- Supplementary Table S2.
bodies, enhanced levels of γIFN [37]. It is becoming evi-
dent that besides sequence, conformation of gluten pep- Clinical Studies in Adults
tides plays an important role in determining peptide immu-
noreactivity. Longer gluten peptides acquire β-sheet con- Kaukinen et al. [47] found no CD symptoms regarding the
formation [37, 42]. levels of small-bowel mucosal CD3+, αβ + and γσ +
Besides qualitative aspects, i.e., structure of prolamin intraepithelial lymphocytes, small-bowel mucosal villous
molecules, and the length and sequence of gluten pep- damage, inflammation or other gastrointestinal CD symptoms
tides, respectively, recent studies also indicate a quanti- in a group of adult CD patients consuming a median of 20 g of
tative aspect of gluten immunoreactivity, i.e., the fact oats per day for up to eight years whereas the consumed oat
that potential immunoreactivity of gluten peptides products come from general stores. Moreover, oats daily
Plant Foods Hum Nutr

consumption had beneficial effects since it provided enhanced and intestinal permeability data after 6, 9 and 15 months of
fiber intake to CD patients consuming oats with respect to double-blind, placebo-controlled, crossover design trials in
those who did not consume oats. No clinical symptoms at 177 children who were more than two years on GFD.
serological (anti-gluten IgA or IgG antibodies and anti- Similarly, no significant differences were found between
transglutaminase 2 antibodies) and histological (small-bowel groups of children consuming oat-enriched GFD with respect
mucosal damage revealing either partial, subtotal or total to children consuming standard GFD regarding serology (an-
villous atrophy) were detected by Aaltonen et al. [48] in a ti-gliadin, anti-tTG2 IgA antibodies) and small bowel mucosal
group of 869 adult celiacs consuming oats as part of GFD architecture by Högberg et al. [58]. Most children did not
for one year when compared to oat non-consuming celiacs. report adverse effects of long-term oat consumption as part
Similarly, Janatuinen et al. [49] found no differences between of GFD in the study by Tapsas et al. [59]; however,
celiacs non-consuming oats and those consuming 46 g per day Tjellström et al. [60] reported enhanced levels of feacal short
for 12 months in histology of duodenal villi. No significant chain fatty acids (SCFAs) including acetic acid and n-butyric
differences in the levels of anti-avenin IgA antibodies were acid in GFD-oats group of children with respect to standard
found in adult groups on GFD who consume oats with non- GFD group which may present a potential risk for develop-
consuming ones [50]. Similarly, Sey et al. [51] found no sig- ment of gut mucosal inflammation.
nificant changes in symptoms scores (symptomatic recur- In conclusion, de Souza et al. [61] and Pinto-Sánchez et al.
rence, serological relapse, histological deterioration) and other [62] provided a meta-analysis of clinical studies dealing with
indicators such as weight, hemoglobin, ferritin, albumin and safety of using pure oats as a part of GFD, and they concluded
tTG levels between CD patients consuming up to 350 g/week that based on the results of 28 clinical studies comprising 661
oats as a part of their GFD (GFD-oat) with whose consuiming patients, no adverse evidence of using pure oats were reported
standard GFD during a 12-week trial. Cooper et al. [52] found regarding intestinal histology, serology, and immunology.
no significant differences in tTG expression, intraepithelial However, the authors pointed at limited quality of clinical
lymphocyte numbers or enterocyte proliferation in 46 CD pa- studies given their limited geographical distribution, and their
tients consuming oat-containing GFD for one year. design indicating that more rigorous, double-blinded and
In contrast, Lundin et al. [8] found enhanced levels of γIFN placebo-controlled studies are needed.
mRNA after a challenge including 50 g of oats per day for
12 weeks in 19 adult CD patients who previously used GFD
only. One of the patients developed partial villous atrophy Conclusions and Future Perspectives
after a challenge. Similarly, Tuire et al. [53] found persistent
intraepithelial lymphocytosis in small bowel while oats con- Oat avenins represent a relatively lower portion of grain stor-
sumption being the only significant factor contributing to this age proteins when compared to wheat gliadins. In addition,
state in adult CD patients although only minor effects of oat avenins reveal relatively lower P + Q content resulting in
intraepithelial lymphocytosis on villous architecture were shorter gluten peptides which can undergo endosomal
found. Similarly, Zanini et al. [54] reported persistent internalisation in dendritic cells (DCs) without any T cell-
intraepithelial lymphocytosis independent on the length of mediated immune response. However, oat avenins reveal
GFD in CD patients. large genetic variability resulting in a variety of gluten pep-
tides with differential immunoreactivity in human gastrointes-
Clinical Studies in Children tinal tract. The majority of avenin-derived gluten peptides
tested are relatively short and do not induce immunogenic
Koskinen et al. [55] studied the effect of 2-year-long diet response; however, a few larger avenin-derived peptides re-
containing oats on 23 children with CD with respect to their vealing immunogenic reactivity were also reported. Based on
levels of jejunal mucosal tTG2-targeted IgA-class autoanti- the results of recent studies, it is becoming evident that some
bodies deposits and found no significant change in the level oat varieties accumulate avenins yielding immunoreactive
of anti-tTG2 autoantibodies in children with CD diagnosis. peptides while many others seem to be safe for CD patients.
Thus, they concluded that consumption of oats does not in- Given the genotypic variability in avenin-derived peptide se-
duce anti-tTG2 autoantibody production at jejunal mucosal quences and their immunoreactivity, urgent need arises to test
level in children with CD. the individual oat varieties used for preparation of oat-derived
In a double-blinded study, Gatti et al. [56] found no clinical foods for GFD. Therefore, the following aims for the future
symptoms regarding gastronintestinal symptoms rate scale research should be outlined:
(GSRS) and intestinal permeability tests (IPT) in a group of 1/ Clinical tests: The aim for clinical researchers
Italian children after a 6-month diet containing pure oats. (immunologists) is to describe clinical symptoms induced by
Similarly, Lionetti et al. [57] found no differences in clinical various avenin-derived gluten peptides on CD patients in or-
(GSRS), serologic (IgA anti-tTG2 and anti-avenin antibodies) der to identify immnuoreactive peptides.
Plant Foods Hum Nutr

2/ Oat breeding: The aim for oat breeders is to select those References
oat genotypes accumulating avenins providing only short glu-
ten peptides with no or minimum immune response in CD 1. Balakireva AV, Zamyatnin AA Jr (2016) Properties of gluten intol-
erance: gluten structure, evolution, pathogenicity and detoxification
patients which would be safe for people suffering from CD.
capabilities. Nutrients 8:644. https://doi.org/10.3390/nu8100644
The basis for achievements of these aims lies in detailed de- 2. Real A, Comino I, de Lorenzo L, Merchán F, Gil-Humanes J,
scription of genetic variability in avenin molecular structure Giménez MJ, López-Casado MA, Torres MI, Cebolla A, Sousa
among oat cultivars. Oat breeding can thus lead to develop- C, Barro F, Pistón F (2012) Molecular and immunological charac-
ment of CD-safe oat varieties with well-characterised avenin terization of gluten proteins isolated from oat cultivars that differ in
toxicity for celiac disease. PLoS One 7:e48365. https://doi.org/10.
composition. Linkage of several prolamin genes to genes con- 1371/journal.pone.0048365
ferring disease resistance could represent a serious obstacle; 3. Sollid LM, Qiao SW, Anderson RP, Gianfrani C, Koning F (2012)
however, precise gene mapping and utilization of modern Nomenclature and listing of celiac disease relevant gluten T-cell
technologies of molecular breeding can resolve this problem. epitopes restricted by HLA-DQ molecules. Immunogenetics 64:
455–460. https://doi.org/10.1007/s00251-0599-z
3/ Oat food processing: Currently, technologies of food
4. Cohen IS, Day AS, Shaoul R (2019) To be oats or not to be? An
processing based on dough fermentation using bacterial and update on the ongoing debate on oats for patients with celiac dis-
fungal proteases can significantly decrease gluten amounts in ease. Front Pediatr 7:384. https://doi.org/10.3389/fped.2019.00384
gluten-containing foods such as wheat dough. For example, 5. De Re V, Magris R, Cannizzaro R (2017) New insights into the
Rizzello et al. [63] demonstrated efficient reduction of gluten pathogenesis of celiac disease. Front Medicine 4:137. https://doi.
org/10.3389/fmed.2017.00137
contents up to 12 mg/kg associated with hydrolysis of albu-
6. Camarca A, Del Mastro A, Gianfrani C (2012) Repertoire of gluten
mins, globulins, glutenins as well as 33-mer peptide in wheat peptides active in celiac disease patients: perspectives for transla-
dough fermented by Lactobacillus (L. brevis, L. tional therapeutic applications. Endocr Metab Immune Disord Drug
sanfranciscensis, L. hilgardii) and Aspergilus (A. niger, A. Ta r g e t s 1 2 : 2 0 7 – 2 1 9 . h t t p s : / / d o i . o r g / 1 0 . 2 1 7 4 /
187153012800493549
oryzae) proteases.
7. Arentz-Hansen H, Fleckenstein B, Molberg Ø, Scott H, Koning F,
4/ Oat purity: Last, but not least, oat contamination with Jung G, Roepstorff P, Lundin KEA, Sollid LM (2004) The molec-
other cereals from Triticeae group often leads to enhanced ular basis for oat intolerance in patients with celiac disease. PLoS
immunoreactivity of oat products. Therefore, the manufac- Med 1(1):e1. https://doi.org/10.1371/journal.pmed.0010001
turers of gluten-free oat products have to adhere strict purity 8. Lundin KEA, Nilsen EM, Scott HG, Løberg EM, Gjøen A, Bratlie
J, Skar V, Mendez E, Løvik A, Kett K (2003) Oats induced villous
protocol to eliminate oat contamination with other cereals
atrophy in celiac disease. Gut 52:1649–1652. https://doi.org/10.
[64]. 1136/gut.52.11.1649
In conclusion, given the genetic variability in avenin 9. Commission Regulation n. 41/2009 concerning the composition
protein sequences and their immunoreactivity in CD pa- and labelling of foodstuffs suitable for people intolerant to gluten.
tients, it is necessary to carefully select and breed new Official J European Union dated 21.01.2009, L16/3. http://eur-lex.
europa.eu/LexUriServ/LexUriServ.do?uri=OJ;L:2009:016:0003:
oat varieties with minimum CD reactivity for GFD. To 0005:EN:PDF
achieve this aim, characterization of variability and immu- 10. Rashid M, Butzner D, Burrows V, Zarkadas M, Case S, Molloy M,
noreactivity of avenin-derived gluten peptides is a neces- Warren R, Pulido O, Switzer C (2007) Consumption of pure oats by
sary prerequisite. In the next step, programmes aimed at individuals with celiac disease: a position statement by the
Canadian celiac association. Can J Gastroenterol 21:649–651.
breeding novel oat varieties carrying only those avenin
https://doi.org/10.1155/2007/340591
alleles providing gluten peptides with no (minimum) im- 11. Joint Australian and New Zealand Food Standards Code. Standard
munoreactivity should result in production of CD-safe oat 1.2.8. 2007. Available at: http://www.foodstandards.gov.au/
foods suitable for GFD. Companies producing oat foods foodstandardscode
suitable for persons suffering from CD have to pay atten- 12. Fric P, Gabrovska D, Nevoral J (2011) Celiac disease, gluten-free
diet, and oats. Nutrition Rev 69:107–115. https://doi.org/10.1111/j.
tion to the origin (genotype) of oats used for food produc- 1753-4887.2010.00368.x
tion. An alternative way can lie in gluten reduction during 13. Comino I, de Lourdes MM, Sousa C (2015) Role of oats in celiac
food processing via bacterial and fungal fermentation. disease. World J Gastroenterol 21:11825–11831. https://doi.org/10.
Considering all these aspects of gluten reduction in oats, 3748/wjg.v21.i41.11825
CD-safe oat represents a matter of control. 14. Eggum BO, Gullord M (1983) The nutritional quality of some oat
varieties cultivated in Norway. Plant Foods Human Nutr 32:67–73
15. Inglett GE, Newman RK (1994) Oat β-glucans amylodextrins: pre-
Acknowledgements The work was supported by an institutional project liminary preparations and and biological properties. Plant Foods
MZe CR RO0418 and by the project QK1810102 of the Ministry of Human Nutr 45:53–61
Agriculture of the Czech Republic. 16. Hardy MY, Tye-Din JA, Stewart JA, Schmitz F, Dudek NL,
Hanchapola I, Purcell AW, Anderson RP (2015) Ingestion of oats
and barley in patients with celiac disease mobilizes cross-reactive T
Compliance with Ethical Standards cells activated by avenin peptides and immuno-dominant hordein
peptides. J Autoimmunity 56:56–65. https://doi.org/10.1016/j.jaut.
Conflict of Interest The authors declare no conflict of interest. 2014.10.003
Plant Foods Hum Nutr

17. Pedó I, Sgarbieri VC, Gutkoski LC (1999) Protein evaluation of 33. Spaenij-Dekking EH, Kooy-Winkelaar EM, Nieuwenhuizen WF,
four oat (Avena sativa L.) cultivars adapted for cultivation in the Drijfhout JW, Koning F (2004) A novel and sensitive method for
south of Brazil. Plant Foods Human Nutr 53:297–304 the detection of T cell stimulatory epitopes of alpha/beta- and gam-
18. Chesnut RS, Shotwell MA, Boyer SK, Larkins BA (1989) Analysis ma-gliadin. Gut 53: 1267–1273. https://doi.org/10.1136/gut.2003.
of avenin proteins and the expression of their mRNAs in developing 037952
oat seeds. Plant Cell 1:913–924 34. Ballabio C, Uberti F, Manferdelli S, Vacca E, Boggini G, Redaelli
19. Londono DM, van’t Westende WPC, Goryunova S, Salentijn EMJ, R, Catassi C, Lionetti E, Penas E, Restani P (2011) Molecular
van den Broeck HC, van der Meer IM, Visser RGF, Gilissen LJWJ, characterization of 36 oat varieties and in vitro assessment of their
Smulders MJM (2013) Avenin diversity analysis of the genus suitability for coeliacs´ diet. J Cereal Sci 54:110–115. https://doi.
Avena (oat). Relevance for people with celiac disease J Cereal Sci org/10.1016/j.jcs.2011.04.004
58: 170–177. http.//dx.doi.org/j.jcs.2013.03.017 35. Benoit L, Masiri J, del Blanco IA, Meshgi M, Samadpour M (2017)
20. Jones DT, Taylor WR, Thornton JM (1992) The rapid generation of Assessment of avenins from different oat varieties using R5-based
mutation data matrices from protein sequences. Computer sandwich ELISA. J Agric Food Chem 65:1467–1472. https://doi.
Applications Biosci 8:275–282. https://doi.org/10.1093/ org/10.1021/acs.jafc.6b05105
bioinformatics/8.3.275 36. Giménez MJ, Real A, Garcia-Molina MD, Sousa C, Barro F (2017)
21. Kumar S, Stecher G, Tamura K (2016) MEGA7: molecular evolu- Characterization of celiac disease related oat proteins: bases for the
tionary genetics analysis version 7.0 for bigger datasets. Mol Biol development of high quality oat varieties suitable for celiac patients.
Evol 33:1870–1874. https://doi.org/10.1093/molbev/msw054 Sci Rep 7:42588–42589. https://doi.org/10.1038/srep42588
22. Anderson OD (2014) The spectrum of major seed storage genes and 37. Comino I, Bernardo D, Bancel E, de Lourdes MM, Sánchez B,
proteins in oats (Avena sativa). PLoS One 9:e83569. https://doi.org/ Barro F, Šuligoj T, Ciclitira PJ, Cebolla A, Knight SC, Branlard
10.1371/journal.pone.0083569 G, Sousa C (2016) Identification and molecular characterization
23. Vader W, Stepniak D, Bunnik EM, Kooy YM, de Haan W, of oat peptides implicated on celiac immune response. Food Nutr
Drijfhout JW, Van Veelen PA, Koning F (2003) Characterization Res 60:30324. https://doi.org/10.3402/fnr.v60.30324
of cereal toxicity for celiac disease patients based on protein ho- 38. Comino I, Real A, de Lorenzo L, Cornell H, López-Casado MÁ,
mology in grains. Gastroenterology 125:1105–1113. https://doi. Barro F, Lorite P, Torres MI, Cebolla Á, Sousa C (2011) Diversity in
org/10.1016/50016-5085/(03)01204-6 oat potential immunogenicity: basis for the selection of oat varieties
24. Kahlenberg F, Sanchez D, Lachman I, Tuckova L, Tlaskalova H, with no toxicity in celiac disease. Gut 60:915–922. https://doi.org/
Mendez E, Mothes T (2006) Monoclonal antibody R5 for detection 10.1136/gut.2010.225268
of putatively celiac-toxic gliadin peptides. Eur Food Res Technol
39. Silano M, Dessi M, De Vincenzi M, Cornell H (2007) In vitro test
222:78–82. https://doi.org/10.1007/s00217-005-0100-4
indicate that certain varieties of oats may be harmful to patients with
25. Valdes I, Garcia E, Llorente M, Mendez E (2003) Innovative ap- coeliac disease. J Gastroenterol Hepatol 22:528–531. https://doi.
proach to low-level gluten determination in foods using a novel
org/10.1111/j.1440-1746.2006.04512.x
sandwich enzyme-linked immunosorbent assay protocol. Eur J
40. Silano M, Di Benedetto R, Maialetti F, De Vincenzi A, Calcaterra
Gastroenterol Hepatol 15:465–474. https://doi.org/10.1097/01.
R, Cornell HJ, De Vincenzi M (2007) Avenins from different cul-
meg.000059119.41030.df
tivars of oats elicit response by celiac peripheral lymphocytes.
26. Hernando A, Mujico JR, Mena MC, Lombardia M, Méndez E
Scand J Gastroenterol 42:1302–1305. https://doi.org/10.1080/
(2008) Measurement of wheat gluten and barley hordeins in con-
00365520701420750
taminated oats from Europe, the United States and Canada by
41. Silano M, Pozo EP, Uberti F, Manferdelli S, Del Pinto T, Felli C,
Sandwich R5 ELISA. Eur J Gastroenterol Hepatol 20:545–554.
Budelli A, Vincentini O, Restani P (2014) Diversity of oat varieties
https://doi.org/10.1097/MEG.0b013e3282f46597
in eliciting the early inflammatory events in celiac disease. Eur J
27. Morón B, Cebolla A, Manyani H, Alvarez-Maqueda M, Megías M,
Nutrition 53:1177–1186. https://doi.org/10.1007/s00394-013-
Thomas Mdel C, López MC, Sousa C (2008) Sensitive detection of
0617-4
cereal fractions that are toxic to celiac disease patients by using
monoclonal antibodies to a main immunogenic wheat peptide. 42. Herrera MG, Pizzuto M, Lonez C, Rott K, Hütten A, Sewald N,
Am J Clin Nutr 87:405–414. https://doi.org/10.1093/ajcn/87.2.405 Ruysschaert JM, Dodero VI (2018) Large supramolecular struc-
28. Halmayr-Jech E, Hammer E, Fielder R, Counts J, Rogers A, tures of 33-mer gliadin peptide activate toll-like receptors in mac-
Cornish M (2012) Characterization of G12 sandwich ELISA, a next rophages. Nanomedicine 14:1417–1427. https://doi.org/10.1016/j.
generation immunoassay for gluten toxicity. J AOAC nano.2018.04.014
INTERNATIONAL 95:372–376. https://doi.org/10.5740/jaoacint. 43. Satheeskumar S, Sharp PJ, Lagudah ES, McIntosh RA, Molnar SJ
sge.halmayr-jech (2011) Genetic association of crown rust resistance gene pc68, stor-
29. Nassef HM, Redondo MCB, Ciclitira PJ, Ellis HJ, Fragoso A, age protein loci, and resistance gene analogues in oats. Genome 54:
O’Sullivan CK (2008) Electrochemical immunosensor for detec- 484–497. https://doi.org/10.1139/G11-014
tion of celiac disease toxic gliadin in foodstuff. Anal Chem 80: 44. Lerner A (2014) The enigma of oats in nutritional therapy for celiac
9265–9271. https://doi.org/10.1021/ac801620j disease. Int. J. Celiac disease 2:110–114. https://doi.org/10.12691/
30. Nassef HM, Civit L, Fragoso A, O’Sullivan CK (2009) ijcd-2-3-1
Amperometric immunosensor for detection of celiac disease toxic 45. Sovová T, Kerrins G, Demnerová K, Ovesná J (2017) Genome
gliadin based on fab fragments. Anal Chem 81: 5299–5307. https:// editing with engineered nucleases in economically important ani-
doi.org/10.1021/ac9005342 mals and plants: state of the art in the research pipeline. Curr Issues
31. Skerritt JH, Hill AS, Andrews JL (2000) Antigenicity of wheat Mol Biol 21:41–62. https://doi.org/10.21775/cimb.021.041
prolamins: detailed epitope analysis using a panel of monoclonal 46. La Vieille S, Pulido OM, Abbott M, Koerner TB, Godefroy S
antibodies. J Cereal Sci 32:259–279. https://doi.org/10.1006/jcrs. (2016) Celiac disease and gluten-free oats: a Canadian position
2000.0316 based on a literature review. Can J Gastroenterol Hepatol, article
32. Mujico JR, Mitea C, Gilissen LJWJ, de Ru A, van Veelen P (2011) ID 18570305:10 pages. https://doi.org/10.1155/2016/1870305
Natural variation in avenin epitopes among oat varieties: implica- 47. Kaukinen K, Collin P, Huhtala H, Mäki M (2013) Long-term con-
tions for celiac disease. J Cereal Sci 54:8–12. https://doi.org/10. sumption of oats in adult celiac disease patients. Nutrients 5:4380–
1016/j.jcs.2010.09.007 4389. https://doi.org/10.3390/nu5114380
Plant Foods Hum Nutr

48. Aaltonen K, Laurikka P, Huhtala H, Mäki M, Kaukinen K, Kurppa Kleon W, Restani P, Brusca I, Budelli A, Gesuita R, Carle F, Catassi
K (2017) The long-term consumption of oats in celiac disease pa- C (2018) Safety of oats in children with celiac disease. J Pediatrics
tients is safe: a large cross-sectional study. Nutrients 9:611. https:// 194:116–122. https://doi.org/10.1016/j.jpeds.2017.10.062
doi.org/10.3390/nu9060611 58. Högberg L, Laurin P, Fälth-Magnusson K, Grant C, Grodzinsky E,
49. Janatuinen EK, Pikkarainen PH, Kemppainen TA, Kosma VM, Jansson G, Ascher H, Browaldth L, Hammersjö JA, Lindberg E,
Jarvinen RMK, Uusitupa MIJ, Julkunen RJK (1995) A comparison Myrdel U, Stenhammar L (2004) Oats to children with newly diag-
of diets with and without oats in adults with celiac disease. New nosed coeliac disease: a randomised double blind study. Gut 53:
Engl J Med 333:1033–1037 649–654. https://doi.org/10.1136/gut.2003.026948
50. Guttormsen V, Løvik A, Bye A, Bratlie J, Møkrid L, Lundin KE 59. Tapsas D, Fälth-Magnusson K, Högberg L, Hammersjö JA, Hollén
(2008) No induction of anti-avenin IgA by oats in adult, diet-treated E (2014) Swedish children with celiac disease comply well with a
coeliac disease. Scand J Gastroenterol 43:161–165. https://doi.org/ gluten-free diet, and most include oats without reporting any ad-
10.1080/00365520701832822 verse effects: a long-term follow-up study. Nutrition Res 34:436–
51. Sey MSL, Parfitt J, Gregor J (2011) Prospective study of clinical 441. https://doi.org/10.1016/j.nutres.2014.04.006
and histological safety of pure and uncontaminated Canadian oats 60. Tjellström B, Stenhammar L, Sundqvist T, Fälth-Magnusson K,
in the management of celiac disease. J Parenteral Enteral Nutrition Hollén E, Magnusson KE, Norin E, Midtvedt T, Högberg L
35:459–464. https://doi.org/10.1177/0148607110387800 (2014) The effects of oats on the function of gut microflora in
52. Cooper SEJ, Kennedy NP, Mohamed BM, Abuzakouk M, Dunne J, children with coeliac disease. Aliment Pharmacol Ther 39:1156–
Byrne G, McDonald G, Davies A, Edwards C, Kelly J, Feighery CF 1160. https://doi.org/10.1111/apt.12707
(2012) Immunological indicators of coeliac disease activity are not 61. de Souza MCP, Deschénes ME, Laurencelle S, Godet P, Roy CC,
altered by long-term oats challenge. Clinical Exp Immunol 171: Djilali-Saiah I (2016) Pure oats as part of the Canadian gluten-free
313–318. https://doi.org/10.1111/cei.12014 diet in celiac disease: the need to revisit the issue. Can J
53. Tuire I, Maria-Leena L, Teea S, Katri H, Jukka P, Paivi S, Heini H, Gastroenterol Hepatol:1576360, 8 pages. https://doi.org/10.1155/
Markku M, Pekka C, Katri K (2012) Persistent duodenal 1576360
intraepithelial lymphocytosis despite a long-term strict gluten-free
62. Pinto-Sánchez MI, Causada-Calo N, Bercik P, Ford AC, Murray
diet in celiac disease. Am J Gastroenterol 107:1563–1569. https://
JA, Armstrong D, Semrad C, Kupfer SS, Alaedini A, Moayyedi P,
doi.org/10.1038/ajg.2012.220
Leffler DA, Verdú EF, Green P (2017) Safety of adding oats to a
54. Zanini B, Marullo M, Villanacci V, Salemme M, Lanzarotto F, Ricci
gluten-free diet for patients with celiac disease: systematic review
C, Lanzini A (2016) Persistent intraepithelial lymphocytosis in ce-
and meta-analysis of clinical and observational studies.
liac patients adhering to gluten-free diet is not abolished despite a
Gastroenterology 153:395–409. e3. https://doi.org/j.gastro.2017.
gluten contamination elimination diet. Nutrients 8:525. https://doi.
04.009
org/10.3390/nu8090525
63. Rizzello CG, De Angelis M, Di Cagno R, Camarca A, Silano M,
55. Koskinen O, Villanen M, Korponay-Szabo I, Lindfors K, Mäki M,
Losito I, De Vincenzi M, De Bari MD, Palmisano F, Maurano F,
Kaukinen K (2009) Oats do not induce systemic or mucosal auto-
Gianfrani C, Gobbetti M (2007) Highly efficient gluten degradation
antibody response in children with celiac disease. J Pediatric
by lactobacilli and fungal proteases during food processing: new
Gastroenterol Nutrition 48:559–565. https://doi.org/10.1097/
perspectives for celiac disease. Appl environ Microbiol 73:4499–
MPG.0b013e3181668635
4507. https://doi.org/10.1128/AEM.00260-07
56. Gatti S, Caporelli N, Galeazzi T, Francavilla R, Barbato M,
Roggero P, Malamisura B, Iacono G, Budeli A, Gesuita R, 64. Allred LK, Kupper C, Iverson G, Perry TB, Smith S, Stephen R
Catassi C, Lionetti E (2013) Oats in the diet of children with celiac (2017) Definition of the purity protocol for producing gluten-free
disease: preliminary results of a double-blind, randomized, oats. Cereal Chem 94:377–379. https://doi.org/10.1094/CCHEM-
placebo-controlled multicenter Italian study. Nutrients 5:4653– 01-17-0017-VO
4664. https://doi.org/10.3390/nu5114653
57. Lionetti E, Gatti S, Galeazzi T, Caporelli N, Francavilla R, Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
Cucchiara S, Roggero P, Malamisura B, Iacono G, Tomarchio S, tional claims in published maps and institutional affiliations.

You might also like