You are on page 1of 13

Special Article

Adverse effects of gluten ingestion and advantages of gluten


withdrawal in nonceliac autoimmune disease
Aaron Lerner, Yehuda Shoenfeld, and Torsten Matthias

In light of the coincident surge in overall gluten intake and the incidence of autoim-
mune diseases, the possible biological adverse effects of gluten were explored.
PubMed, MEDLINE, and the Cochrane Library databases were screened for reports

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


published between 1964 and 2016 regarding the adverse effects of gluten as well
as the effects of a gluten-free diet on autoimmune diseases. In vitro and in vivo
studies describing gluten intake in animal models or cell lines and gluten-free diets
in human autoimmune diseases were reviewed. Multiple detrimental aspects of glu-
ten affect human health, including gluten-dependent digestive and extradigestive
manifestations mediated by potentially immunological or toxic reactions that in-
duce gastrointestinal inadequacy. Gluten affects the microbiome and increases in-
testinal permeability. It boosts oxidative stress and affects epigenetic behavior. It is
also immunogenic, cytotoxic, and proinflammatory. Gluten intake increases apo-
ptosis and decreases cell viability and differentiation. In certain nonceliac autoim-
mune diseases, gluten-free diets may help curtail the adverse effects of gluten.
Additional in vivo studies are needed to unravel the puzzle of gluten effects in
humans and to explore the potential beneficial effects of gluten-free diets in auto-
immune diseases.

INTRODUCTION Increased consumption of gluten

Gluten is the main protein in wheat, constituting ap- Wheat is an important food in human nutrition, and
proximately 80% of the total proteins in this grain. The wheat- or gluten-containing foods are common in
remaining 20%—albumins and globulins; multiple Western diets. Wheat grain is easy to stockpile and con-
enzymes such as b-amylases, uridine diphosphate venient to grind into flour, which improves its texture,
glucose pyrophosphorylases, peroxidases, and thiore- and palatability. According to the Food and Agriculture
doxins; amylase-trypsin inhibitors; puroindolines; heat- Organization of the United Nations, worldwide produc-
shock proteins; and proteins involved in response to tion of wheat increased from 711.4 million metric tons
stress— are outside the scope of this review. Wheat also in 2013 to 750.1 million metric tons in 2017 (as of
contains nonprotein components like fermentable, October 5, 2017).2 Gluten is a major food additive in
oligo-, di-, and monosaccharides and polyols the processed food industry, with a net increase in usage
(FODMAPs). Not surprisingly, gluten peptides are also of 1.8 6 0.4% per year reported over the last 30 years1
contained in other nonwheat cereals like barley, rye, Currently, both the prevalence and the incidence of
and oats.1 gluten-related disorders are surging in many countries

Affiliation: A. Lerner is with the B. Rappaport School of Medicine, Technion–Israel Institute of Technology, Haifa, Israel. Y. Shoenfeld is with
the Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center–Tel Hashomer, Ramat Gan, Israel, and the Sackler Faculty of
Medicine, Tel-Aviv University, Tel-Aviv, Israel. A. Lerner and T. Matthias are with the AESKU.KIPP Institute, Wendelsheim, Germany
Correspondence: A. Lerner, AESKU.KIPP Institute, Mikroforum Ring 2, Wendelsheim 55234, Germany. Email: aaronlerner1948@gmail.com.
Key words: adverse effect, autoimmunity, biological effects, celiac disease, gluten-free diet.
C The Author(s) 2017. Published by Oxford University Press on behalf of the International Life Sciences Institute.
V
All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.

doi: 10.1093/nutrit/nux054
1046 Nutrition ReviewsV Vol. 75(12):1046–1058
R
worldwide.3–7 The reasons most frequently suggested lines, and (3) effects of gluten-free diets on nonceliac
include the spread of the Western diet,1 the increased autoimmune diseases in humans.
consumption of gluten-containing diets,8 the gradual Research studies, reviews, and case–control series
shift toward increased production and consumption of were included, while case reports were excluded. The liter-
wheat, which is replacing the traditional food of rice in ature search was performed using the PubMed,
Asia and Africa,9 and the evolutionary development of MEDLINE, Embase, Scopus, and Cochrane Database of
wheat with higher gluten content and cytotoxic proper- Systematic Reviews databases to identify the most relevant
ties.10,11 Even advanced food-processing technology, information.
which reduces dough fermentation time, can increase The following search algorithm was used (“gluten
gluten concentration in commercial bakery prod- side effect” or “gluten adverse effects” or “gluten
ucts.10,12 It is estimated that, in the 20th century, global treatment” or “gluten therapy” or “gluten application”)
wheat output increased 5-fold, with most of the increase AND (“human” or “animal” or “cell-line”) AND

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


occurring after 1955.4 In parallel, the prevalence of (“gluten”) or (“gliadin”) AND (“autoimmune disease”).
celiac disease increased 5-fold in the United States be- Type 1 diabetes, multiple sclerosis, psoriasis, rheu-
tween 1974 and 1989.13 On the other hand, in a 2014 matoid arthritis, autoimmune hepatitis, celiac hepatitis,
US Consumer Reports survey, 63% of responders felt autoimmune thyroiditis, and autoimmune diseases
that abstaining from bread and other gluten-containing were searched separately for gluten or gluten-free diets.
diets would enhance their physical or mental health. Additional studies were identified by examining the ref-
Moreover, in a 2015 Gallup poll, 21% of Americans dis- erence list of the retrieved articles. The search was lim-
closed they tried to follow a gluten-free diet.14 ited to articles published in English. Altogether, 251
publications were identified. Relevant articles were se-
Increased frequency of autoimmune disease lected for full-text review on the basis of screened titles
and abstracts. Since primary data was not collected, eth-
Increasing evidence demonstrates a continuous rise in ical approval was not mandatory.
the incidence of autoimmune diseases.3 The incidence of
rheumatic, endocrinological, gastrointestinal, and neuro- DIRECT IMMUNE EFFECTS OF GLUTEN IN
logical autoimmune diseases has increased annually, at ANIMAL STUDIES
rates ranging from 3.7% to 7.1%, between 1985 and
2015.3 These observations, together with the fact that only Direct effects of gluten on dendritic cells and innate
1% to 3% of the 30% of the general population that carries immune systems in mice
the human leukocyte antigen DQ 2/8 allele will develop
celiac disease, strengthen the hypothesis that the environ- The innate immune system plays a critical role in oral tol-
ment plays a more important role than genetics, in the erance to nutritional antigens. In experiments with
development of autoimmune diseases, in the last BALB/c and nonobese diabetic (NOD) mice, gluten-free
decades.3,4,15 The increased incidence of celiac disease has diets increased both the percentage of macrophages in the
been linked to increased gluten consumption.4,15 However, spleen of BALB/c mice and the percentage of CD11cþ
some evidence indicates that gluten might be related to the dendritic cells in the spleen of BALB/c and NOD mice.
increased incidence of other, nonceliac autoimmune dis- Moreover, gluten-free diets increased the percentage of
eases. The parallel surge in gluten consumption3,15 and the CD103þ dendritic cells in BALB/c mice and decreased the
increased incidence of autoimmune diseases prompted a percentage of CD11bþ dendritic cells in their mesenteric
review of the current knowledge about the disadvantages lymph nodes. Gluten-free diets in BALB/c mice attenuated
of dietary gluten and the role of gluten-free diets in the nu- the exposure of dendritic cells to CD40, CCR7, and major
tritional therapy of autoimmune diseases. histocompatibility complex II in pancreatic lymph nodes.
These results help to explain the low incidence of diabetes
METHODS in gluten-free NOD mice, in which gluten might play a di-
abetogenic role. This pathway might be essential in the
The first part of this review addresses the effects of glu- progression of type 1 diabetes (T1D), celiac disease, and
ten on cell lines of human origin in vitro and on animal nonceliac gluten sensitivity.16
models in vivo, since in vivo studies in humans are
scarce. The second part addresses the effects of gluten- Direct effects of gluten on cytokine production and
free diets in various autoimmune diseases. The litera- cytotoxicity of natural killer cells
ture search covered the period 1964–2016 and included
studies that describe the following: (1) effects of gluten Larsen et al.17 have shown that gluten-containing diets
intake by animals, (2) in vitro effects of gluten on cell drive an increased expression and cytotoxicity of

Nutrition ReviewsV Vol. 75(12):1046–1058


R
1047
natural killer cells toward pancreatic beta-cell line intestinal autoimmunity. In studies of enteric and pan-
MIN6 cells, higher expression NKG2D and CD71 on creatic islets in BALB/c and NOD mice, gluten-free
NKp46(þ) cells in all lymphoid organs of BALB/c and diets decreased the expression of natural killer group
NOD mice. Gluten ingestion augments the secretion of 2D and its ligands.21 This immunological response may
both IFN-c and IL-6 to a greater degree in BALB/c and lower T1D incidence associated with gluten-free diets
NOD mice than in mice fed gluten-free diets, suggest- in animals and, potentially, in humans.
ing that gluten intake induces murine natural killer cell
activity toward pancreatic beta cells. This may reinforce
the progression of T1D and help explain the increased Induction of inflammation through Toll-like receptor 4
incidence of disease associated with gluten consump-
tion in NOD mice.17 Animal and cell culture models have shown that gluten
peptides can induce inflammation by binding Toll-like

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


Direct effects of gluten on the Th17 pathway receptor 4, thereby inducing the expression of proin-
and T regulatory cells flammatory cytokines such as IL-1, IL-6, and tumor ne-
crosis factor.22,23 Although these findings must still be
Cytokine-secreting Th17 cells are associated with auto- corroborated in humans, they suggest an intriguing hy-
immunity. The Th17 profile is substantially increased in pothesis, given the current trend toward increased use
the pancreatic lymph nodes of BALB/c gluten-fed mice, of gluten-free diets. Table 116–18,20,21,24–38 summarizes
parallel to the increased populations of CD4þ, additional studies reporting in vivo and in vitro direct
CD45RBhighþ, and CD103þ T cells. This indicates that effects of gluten intake on the immune system. Taken
gluten intake impacts multiple subsets of T regulatory together, the animal studies support the involvement of
cells as well as Th17 cells in the mucosal lymphoid gluten peptides in the innate and the regulatory im-
tissue.18 mune systems, on the Th17 and other proinflammatory
pathways and neutrophil migration. There are some
Closure of ATP-sensitive potassium channels indications that gluten may be involved in the induction
and induction of insulin release of diabetes and autoimmune disorders. The following
section on adverse effects of gluten in humans might
The incidence of diabetes is reduced by a gluten-free shed additional light on the subject.
diet in the NOD murine model.19 Gluten peptides, such
as gliadin, can cross the intestinal barrier and may di-
rectly affect the beta cells of the endocrine pancreas. In ADVERSE EFFECTS OF GLUTEN ON HUMAN HEALTH
INS-1E cells and in isolated rat islets, the application of
gliadin peptides induced a dose-related surge in insulin The role of gluten in celiac disease is well established
delivery and decreased electrical currents through ATP- and will not be described in this review. Yet gluten also
sensitive potassium channels.19 This data may indicate plays a role, though indirectly, in other nonceliac health
gliadin fragments as modulators responsible for the conditions.
activation of beta cells activity in the late stages of T1D. Gliadin is an ideal substrate for endogenous tissue
transglutaminase and exogenous microbial transgluta-
Induction of neutrophil migration minase, both of which belong to the extended transglu-
taminase family.39 Gluten is abundant in glutamine and
Gliadin induces the secretion of interleukin (IL) 8, a lysine and thus is a very attractive substrate for post-
powerful neutrophil-activating and chemoattractant translational modification of protein by both endoge-
chemokine. Recently, gliadin was shown to exhibit neu- nous tissue transglutaminase and exogenous microbial
trophil chemoattractant capabilities similar to those of transglutaminase. Both enzymes are able to deamidate
the classical neutrophil chemoattractant, N-formyl- or cross-link gluten peptides to numerous other pepti-
methionyl-leucyl-phenylalanine (fMet-Leu-Phe), and des, thus turning naive, self-proteins into immunogenic
likewise uses the formyl peptide receptor 1 to induce ones.40–44 Importantly, luminal microbial transglutami-
migration.20 nase, derived from processed food or from the micro-
biome or, more specifically, from the dysbiome
Induction of expression of the activating receptor (microbial imbalance), is a major cause of post-transla-
natural killer group 2D and its ligands tional modification of gliadin or other naive proteins.45
Recently, it was hypothesized that the luminal transglu-
Enteric expression of the activating receptor natural taminases, which originate in the dysbiome, are poten-
killer group 2D (CD314) and its ligands is a marker of tial drivers of end-organ or systemic autoimmunity.46

1048 Nutrition ReviewsV Vol. 75(12):1046–1058


R
Table 1 Direct in vivo and in vitro effects of gluten intake on the immune system
Reference Study Cell type or Effects of gluten intake
type animal model
Thomas et al. (2006),24 In vitro Macrophages Increases proinflammatory cytokine and nitric oxide
Tuckova et al. (2000),25 production
Tuckova (2002)26
Nikulina et al. (2004),27 In vitro Dendritic cells Upregulates MHC II, costimulatory molecules, Toll-like re-
Palova-Jelinkova et al. (2005),28 ceptor, cytokine, and chemokine production
Ciccocioppo et al. (2008)29
Larsen et al. (2014)17 In vitro Lymphoid organ Increases expression of NKG2D and CD71 on NKp46þ
cells
Gujral et al. (2015)30 In vitro Caco-2 Increases intestinal permeability, TNF-a, and IL-1b
production
Flohe et al. (2003),31 In vivo NOD mice Modifies Th1/Th2 intestinal cytokines

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


Alam et al. (2010)32
Alam et al. (2010)32 In vivo NOD mice Increases counts of activated intestinal CD4þ T cells, den-
dritic cells, and Th17 cells
Scott et al. (2002)33 In vivo BB rats Modifies Th1 intestinal and MLN cytokines
Scott et al. (1997)34 In vivo BB rats Alters Th1 cytokine pattern in islet infiltrate
Antvorskov et al. (2012)18 In vivo BALB/c mice Induces proportional changes in regulatory T-cell subsets
Antvorskov et al. (2012)18 In vivo BALB/c mice Increases Th17 cell counts in peripheral lymph nodes
Antvorskov et al. (2013),35 In vivo BALB/c mice Alters inflammatory cytokine pattern in FOX3 and
Bernardo et al. (2007) 36
FOXP3þ T cells, activates innate immune response
Antvorskov et al. (2012)18 In vivo BALB/c mice Increases Th17 cell counts, increases proportions of
CD4þCD45RBhighþ cells, increases CD103þ T-cell
counts
Larsen et al. (2015)16 In vivo BALB/c and NOD mice Modulates dendritic cells and innate immune subsets in
mice
Larsen et al. (2014)17 In vivo BALB/c and NOD mice Increases natural killer cell cytotoxicity and cytokine se-
cretion of IFN-c and IL-6
Adlercreutz et al. (2014)21 In vivo BALB/c and NOD mice Induces expression of NKG2D and ligands
Palova-Jelinkova et al. (2013)37 In vivo NLRP/ ASC/ KO mice Induces IL-1b production in bone-marrow-derived den-
dritic cells
Lammers et al. (2015)20 In vivo C57BL/6 and Lys-GFP mice Induces neutrophil migration
Stepankova et al. (1996)38 In vivo AVN Wistar rats Increases counts of pathogenic intraepithelial
lymphocytes
Abbreviations: IL, interleukin; IFN, interferon; KO, knockout; MHC, major histocompatibility complex; MLN, mesenteric lymph node; Th,
T helper; TNF, tumor necrosis factor.

Immunogenicity of gluten immunoglobulin G (IgG), immunoglobulin A (IgA),


and IgM antibody responses than products containing
Antigliadin antibodies are age dependent and are found raw wheat. It is suggested that industrial processing of
in the normal population well as in patients with intesti- wheat may enhance the immunogenicity of the protein
nal and extraintestinal diseases.47–50 Antigliadin anti- (gluten).53
bodies may represent an epiphenomenon whereby the Microbial transglutaminase docked by gluten pepti-
immune system reacts against gluten peptides. Even in des is immunogenic in celiac disease.40 Microbial
the PreventCD intervention study, 30% of healthy transglutaminase–modified gluten proteins react with
infants produced antibodies against gluten/gliadin with- IgA antigliadin antibodies in the sera of patients with
out anti–tissue transglutaminase antibodies shortly after celiac disease. Microbial transglutaminase–treated glu-
gluten introduction.51 In patients with rheumatoid ar- ten peptides cultured with intestinal biopsies from
thritis, a non–gluten-related condition, the intestinal patients with celiac disease induced a substantial release
fluid and sera were significantly enriched with immu- of interferon-! accompanied by a surge in anti–tissue
noglobulin M (IgM) antibodies against gliadin.44,52 transglutaminase and antiendomysial autoantibodies,
When the immunogenicity of processed food products suggesting that those gluten peptides are proinflamma-
vs their raw constituents was checked in normal sera, tory and immunogenic, at least ex vivo. Interestingly, in
titers of the specific corresponding immunoglobulins sera from celiac patients, IgA antibodies were higher
were significantly higher against the processed prod- against wheat from microbial-transglutaminase-treated
ucts. When pizza, pasta, cereals, or bakery products breads than against gluten-free breads not treated with
from supermarket shelves were checked, products con- microbial transglutaminase. Moreover, microbial-
taining processed wheat provoked much higher transglutaminase-deamidated gluten peptides react with

Nutrition ReviewsV Vol. 75(12):1046–1058


R
1049
gluten-selected T cells, thus boosting the immunogenic- their capacity as substrates for enzymatic post transla-
ity of gluten.40 It appears that commercially available tional modification of proteins their immunogenicity
products contain certain quantities of microbial trans- (whether alone or complexed), their ability to reduce
glutaminase, implying that microbial transglutaminase cell viability, their necrotogenic and apoptotic proper-
used during food processing is eaten by consumers and ties, and their ability to breach tight-junction integrity.
eventually finds its way to the gut lumen.54
Effects of gluten on the microbiome
Immunogenicity of gliadin when docked on tissue
transglutaminase or microbial transglutaminase Human beings are host to a multiform but host-specific
gut microbial population along the proximal-to-distal
Even in celiac disease, neoepitopes generated by the axis of the intestine. Analysis of the microbiota in the
docking of gliadin onto tissue transglutaminase and large intestine has shown that nutrition affects the lumi-

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


microbial transglutaminase are more immunogenic than nal bacteria, and together affect the host’s well-being.
undocked gliadin. Compared with conventional tissue Does gluten affect the intestinal ecosystem? The answer
transglutaminase isotypes, the tissue transglutaminase is yes.
neoepitope IgA þ IgG isotype (detected by AESKULISA, When NOD mice were fed a gluten-free diet, num-
CeliCheck New Generation, Wendelsheim, Germany) bers of cecal microbes and gram-positive bacteria were
has a higher optical density value, more accurately lower than those in mice fed a standard gluten-
reflects intestinal injury, has higher sensitivity and specif- containing diet.67,68 Recently, Antvorskov et al.67 and
icity, and targets different autoantigens.55,56 The same Marietta et al.69 showed that gluten-containing diets in-
was found for the microbial transglutaminase creased the numbers of Bifidobacterium, Tannerella,
neoepitope.57 and Barnesiella species in the intestinal flora of NOD
mice, whereas gluten-free diets resulted in increased
Pathogenicity of gluten in HCT116 cells counts of Akkermansia species. It appears that the
microbiome is altered in celiac disease, as a richer,
When applied to HCT116 cells, gliadin reduced cell via- more diverse microbiome has been observed.70 As
bility by 50% (P < 0.05), induced lactic dehydrogenase stated recently by Sanz,70 “disease-associated alterations
secretion over time (P < 0.01), and increased apoptosis in microbiome-derived metabolites could not only re-
significantly (A.L. and T.M., unpublished data, 2016). flect intestinal dysbiosis but also contribute to dysfunc-
These effects of gliadin are supported by multiple stud- tion of the immunoregulatory mechanism.” However,
ies in cell lines of human origin. Decreased viability,58,59 in celiac disease, the situation is more complex, since
necrosis and/or lactic dehydrogenase release,59 and in- crosstalk between the host genome and members of the
duction of apoptosis59–61 have been described. microbiota related to the development of celiac disease
was recently observed.44,71
Ability of gliadin to increase intestinal permeability The precise pathomechanisms that determine how
gluten impacts the intestinal ecosystem are not well un-
Tight-junction integrity is breached in multiple autoim- derstood, but several potential mechanisms, described
mune diseases, including celiac disease.62,63 Numerous below, have been offered.
in vitro, ex vivo, and in vivo studies have confirmed that Gluten-induced dysbiosis is accompanied by a
gliadin, the main antigen of gluten, increases intestinal breach in the tight-junction barrier and the induction
permeability.64,65 After binding to its chemokine receptor of several inflammation-associated microRNAs.72 In
CXCR3 on epithelial cells, gliadin activates the MyD88- addition, the crosstalk between the enteric microbiota
pathway, resulting in the release of zonulin. Gliadin has and the intestinal epithelium is mediated by receptors
agglutinating activity, polymerizes soluble G- and F- shared with the innate immune system. Thus, gluten-
actins, inhibits cell proliferation, is proapoptotic, induced dysbiosis may contribute to the activation of
decreases redox potential, affects the rearrangement of this inflammatory pathway. Patients with celiac disease
the enterocyte cytoskeleton, and induces zonulin secre- are known to have fewer beneficial gut species and a
tion, resulting in increased gut leakage.63,65 greater number of potentially pathogenic ones com-
Most recently, studies of epithelial transit of immu- pared with healthy controls. Gluten-free diets reduce
nogenic and toxic gliadin peptides have shown en- this dysbiosis, although it is not eliminated.73
hanced luminal degradation of the peptides, Additionally, dietary factors, such as meat-containing
highlighting the importance of the epithelial barrier or vegetarian diets or the presence or absence of gluten
against shorter gliadin peptides.66 In summary, gluten in the diet, can affect a number of taxa in the lumen
and gliadin induce multiple adverse effects, owing to and influence multiple bacterial pathways, especially

1050 Nutrition ReviewsV Vol. 75(12):1046–1058


R
those involved specifically in carbohydrate and starch observed.78,79 Several aspects of the relation between
metabolism. Variation in the diet, including the intro- gliadins and epigenetics were observed not only in ce-
duction of gluten-free diets, could affect the composi- liac disease but also in nonceliac cells like SH-SY5Y, a
tion of the microbiome, even in normal volunteers.74 cell line of neuroblastoma origin, and Caco-2 cells.79
There are also other potential mechanisms by which These results indicate the potential of gliadin peptides
gluten might impact luminal conditions. Compared with to trigger antioxidant and epigenetic alterations that
healthy controls, untreated and treated patients with celiac may be crucial during the postnatal transition from
disease showed evidence of gluten-induced dysbiosis at the breastfeeding to gluten consumption.78,79 Most recently,
duodenal and/or the fecal level.74,75 The ratio of protective the involvement of the proinflammatory cytokine inter-
antiinflammatory microbes such as Lactobacillus and feron-c and the microbiota metabolome in switching
Bifidobacterium to potentially harmful microbes like epigenetic behavior was described in celiac disease.81
Bacteroides and Enterobacteriaceae was lower in untreated Reduced antioxidant capacity and the epigenetic conse-

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


children than in children treated with a gluten-free diet. quences of the gliadin-induced opioid activities may
Similar to observations in dysbiotic microbiota, serum, fe- predispose genetically susceptible individuals to inflam-
cal, and urinary metabolomes from patients with celiac dis- mation or autoimmunity, partially explaining the bene-
ease showed abnormal levels of free amino acids and fits of gluten-free diets. In a more holistic way, the
volatile organic substances that may convey messages to oxidative stress, gene expression, and inflammation in-
the mucosal wall. There is no consensus across studies with duced by gluten-derived peptides during progression of
regard to the specific microbes and/or metabolites in indi- the autoimmune and inflammatory cascade are closely
viduals with untreated or treated celiac disease.75 Finally, it related and interconnected.
should be emphasized gluten affects the microbiome, but
the microbiome also affects gluten and might play a role in Effects of gluten on cellular metabolism
the breakdown and immunogenicity of gluten.76
In the 1990s, wheat-derived peptides were shown to af-
Induction of oxidative stress by gluten fect cell metabolism in Caco-2 cell lines. One study
reported that cell proliferation was inhibited by 50%,
Hudson et al.77 were the first to report the suppressed colony-forming ability was decreased by 20%, and alka-
growth and morphological alterations induced by appli- line phosphatase activity during cell differentiation was
cation of gliadin peptides to cell lines of human origin. decreased.82 Furthermore, peptic/tryptic digests from
Gliadin induces agglutination of K562 cells, decreases wheat inhibited DNA and RNA synthesis by almost
F-actin expression in intestine 407 cells, attenuates cell 80% and glycoprotein synthesis by close to 60%.83
viability and growth, is proapoptotic and affects redox Apoptosis induced by gliadin peptides was demon-
homeostasis in Caco-2 cells, alters the morphology of strated in several cell lines, including Caco-2, LoVo,
LoVo cells and of cells in 2- and 3-dimensional cultures, Hep-2, and MRC-5.59,61,84 The effects of prolamin-
and induces restructuring of the cytoskeleton in intesti- derived antinutritional gliadin peptides, which include
nal epithelial cell line 6.59 Glutathione is a crucial low- antiproliferation, antidifferentiation, cellular agglutina-
molecular-weight molecule that affects redox status. Its tion, and synthesis of nucleic acids, can disturb cellular
depletion perturbs cell homeostasis, triggering cytotox- metabolism and intestinal recovery in various enteric
icity. In fact, the oxidative stress induced by gliadin was inflammatory conditions and gluten-related diseases.
demonstrated in multiple cell lines, including Caco-2,
HT29, SH-SY5Y, T84, and LoVo, and was reviewed ex- Effects of gluten on cognitive function
tensively.59,78–80 The compromised antioxidant defense
mechanisms may exacerbate mucosal inflammation, po- Gluten initiates an immune reaction and decomposes,
tentiating tissue damage and delaying the return of epi- during luminal digestion, to peptides with opioid activ-
thelial cell layer integrity, not only in celiac disease but ity, which has multiple consequences on mental func-
also in other inflammatory conditions. tion and behavior in humans.85 Gluten-derived
peptides that exhibit strong opioid activities are called
Effects of gluten on epigenetic programming exorphins. Exorphin receptors are widely scattered
throughout the body, at locations that include the gut,
Epigenetic programming, including methylation and the brain, and the nervous system. If the intestinal and
histone modification, is affected by numerous environ- blood–brain barriers are disrupted by stress, dietary
mental factors, including nutrients.78,79 Altered expres- components,40,63 alcohol, or over-the-counter drugs,
sion of genes involving methylation homeostasis, gluten-derived exorphins can impact cognitive
induced by gluten-derived peptides, was recently function.85

Nutrition ReviewsV Vol. 75(12):1046–1058


R
1051
The ability of gluten to affect mental function is not
merely a hypothesis. Multiple studies have shown a high
prevalence of antigliadin antibodies in patients with
schizophrenia.86 Patients with autistic spectrum disor-
ders are affected by gluten intake. Gluten-free diets are
efficient in modulating both gastrointestinal symptoms
and behavior in patients with autistic spectrum disor-
der.87 Various risk factors for the progression of schizo-
phrenia can be related to a common pathway in the
intestinal tract. It is increasingly recognized that bidi-
rectional communication, involving neural, hormonal,
and immunological routes, exists between the brain and

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


the gut. An increased incidence of gastrointestinal bar-
rier dysfunction, food antigen sensitivity, inflammation,
and the metabolic syndrome is seen in patients with
schizophrenia.86 These findings may be influenced by
the composition of the gut microbiota. A significant
subgroup of patients may benefit from adherence to a
gluten-free diet. Antimicrobial agents and probiotics
have therapeutic potential to attenuate the metabolic ab-
normalities and immune dysfunction encountered in
patients with schizophrenia.88 A form of gluten Figure 1 Autoimmune diseases that improve following gluten
psychosis—a clinical psychosis clearly related to gluten withdrawal. Abbreviations: AT, autoimmune thyroiditis; AH, auto-
immune hepatitis; MS, multiple sclerosis; PS, psoriasis; RA, rheuma-
consumption, which completely resolved after a gluten-
toid arthritis; T1D, type 1 diabetes.
free diet was implemented—has been described.89 Food
proteins, including gluten, affect the rates of synthesis of
non–human leukocyte antigen genes, it is conceivable that
amino acids and brain neurotransmitters in rats.90
this genetic load transfers some gluten-dependent toxicity
Prolonged gluten intake impairs cognitive performance
to other autoimmune diseases.99–104 Since celiac disease
in elderly patients with celiac disease,91 many of whom
and other autoimmune diseases share common genetic,
benefit from gluten-free diets.92 Multiple behavioral and
environmental, and immunological pathways, it would be
peripheral or central neurological manifestations have
interesting to investigate the possible therapeutic effects of
been described in patients with celiac disease, some of
gluten-free diets in patients with nonceliac autoimmune
whom respond to gluten-free diets.93–96
conditions. At least in celiac disease, the additive risk of
Gluten exorphin B5 is an opioid peptide identified
subsequent autoimmune disease was decreased in patients
after wheat gluten digestion. It stimulates prolactin se-
who adhered to a gluten-free diet vs those who did not.105
cretion and affects brain neurotransmitter discharge,
It was concluded that a gluten-free diet has a protective ef-
even without crossing the blood–brain barrier, in rats.97
fect. The effects of gluten in non–gluten-sensitive popula-
The scientific and medical communities are wit-
tions were recently reviewed.106,107 The protective effect of
nessing a major era in which the relationship between
gluten withdrawal has been observed in several autoim-
the gut and the remote organs, including the gut–brain
mune diseases (Figure 1). Moreover, the ingestion of glu-
axis, is under intense investigation.98 The beneficial or
ten seems to be related to disease progression. It should be
partial response to gluten withdrawal suggests some de-
stressed, however, that the Speculation about the benefits
gree of cause-and-effect relationship between gluten
of gluten-free diets is based on a limited number of clinical
and neurological manifestations.
trials that investigated gluten-free diets as either a causal
To provide support that gluten intake may have ad-
factor in the pathogenesis of disease or in association with
verse effects in patients with nonceliac autoimmune disease,
the improvement of symptoms.
the next section describes various autoimmune diseases in
which gluten-free diets had some beneficial effect.
Type 1 diabetes mellitus
ROLE OF GLUTEN-FREE DIET IN NONCELIAC
AUTOIMMUNE DISEASES Multiple findings suggest a connection between T1D and
gluten or celiac disease: shared genes, coexistence of TID
Since celiac disease and many autoimmune diseases share and celiac disease in some patients, increased celiac disease
some human leukocyte antigen genes and multiple prevalence in patients with T1D, increased selective T-cell

1052 Nutrition ReviewsV Vol. 75(12):1046–1058


R
response and high concentration of wheat antibodies in the partial response of celiac disease–associated rheu-
both T1D and celiac disease, the dependency of animal matic manifestations to gluten-free diets is logical.
models of T1D on diet, with gluten being a major potent Additionally, there is a protective effect of gluten with-
diabetes-inducing protein, and, recently, the identification drawal on the cumulative incidence of additional auto-
of T1D-related proteins from wheat, showing that cDNA immune diseases in patients with celiac disease.78,105 In
clones of diverse wheat storage globulins were associated fact, dietary modifications, like gluten-free vegan diets,
with islet damage.108,109 Moreover, a partial depletion of had clinical benefits in certain patients with rheumatoid
regulatory T cells before gliadin sensitization induced se- arthritis that may result from reduced immunoreactiv-
vere insulitis in a mice model. The presence of gliadin- ity to food antigens that are eliminated by a restrictive
responsive T cells in the pancreatic lymph nodes of mice diet.115 A gluten-free vegan diet in patients with rheu-
that develop insulitis suggests that gliadin-specific T-cell matoid arthritis drives changes that are potentially athe-
clones have a role in the induction of insulitis in this ani- roprotective and antiinflammatory.116 Of note, human

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


mal model.110 leukocyte antigen DQ2/8-restricted gluten-specific T
Gluten-free diets delayed and prevented diabetes in cells were noticed to migrate from the enteric mucosa
NOD mice that had never ingested gluten.111 The intesti- into peripheral blood upon gluten challenge, thus indi-
nal immune system plays a primary role in the pathogene- cating an additional mechanism of articular pathology
sis of T1D, since diabetogenic T cells are initially primed in celiac disease and suggesting a basis for the benefit of
locally in the gut, islet-infiltrating T cells express gut- gluten-free diets.
associated homing receptors, and mesenteric lymphocytes
passively transfer diabetes from NOD mice to mice with
NOD or severe combined immunodeficiency.67 During Multiple sclerosis
infancy, the amount, timing, and mode of gluten intro-
duction have been shown to affect the diabetogenic poten- Numerous neurological manifestations that mimic mul-
tial of gluten, and it is now suggested that a gluten-free tiple sclerosis have been described in patients with ce-
diet may preserve beta cell function.67 A subset of patients liac disease.93,94 A certain percentage of patients with
with T1D had an abnormal immune reaction to gluten, multiple sclerosis show high levels of IgA anti–tissue
manifested by a positive reaction to rectal gluten challenge transglutaminase autoantibodies.117 It has been recom-
that resulted in lymphocyte infiltration of the rectal mu- mended that gluten-free diets be considered in patients
cosa, increased proliferative T-cell response to wheat pro- who test positive for antigliadin antibodies. More re-
teins, with production of proinflammatory cytokines, and cently, an increased prevalence of antibodies associated
an elevated T-cell response to gluten in one-fourth of with celiac disease was found in a population of patients
patients with newly diagnosed T1D. All of these effects oc- with relapsing-remitting multiple sclerosis (odd ratio,
curred in patients without celiac disease–associated hu- 5.33),118 and 11% of that population was diagnosed as
man leukocyte antigen haplotype, suggesting a direct, having celiac disease. This is 5 to 10 times greater than
diabetes-specific effect of gluten.67 the prevalence in the normal population. Furthermore,
Notably, the benefits of a gluten-free diet in 32% of first-degree relatives of this population had posi-
patients with symptomatic celiac disease and T1D are tive serology for celiac disease. More relevant to the pre-
supported by the literature, though it is unclear whether sent review, however, is the finding that “the response
these benefits occur in asymptomatic patients. In a 6- of the gluten-free diets was excellent in all of them, both
month study of a gluten-free diet in individuals at high from the digestive and the neurologic point of view in
risk for T1D, autoantibody levels did not change, but the average follow-up period of three years long.”
insulin secretion was reduced.112 Other studies did not Several other small series and case reports observed
substantiate the benefit of gluten-free diets.113,114 Taken clinical improvement in patients on gluten-free diets,
together, gluten consumption may play a substantial but further research is needed to evaluate the response
role in the progression of T1D, but additional studies of patients with multiple sclerosis to nutritional
are warranted to explore whether gluten withdrawal changes.119,120 Many immunopathogenic pathways and
could prevent disease in susceptible individuals or be various celiac disease–associated antibodies have been
applied in newly diagnosed patients to stop or attenuate described. These antibodies can cross the blood–brain
disease progression. barrier and deposit at the level of the Purkinje cells,
producing a remarkable inflammatory reaction fol-
Rheumatoid arthritis lowed by neuronal damage and cerebellar atrophy.118
The isoenzyme of tissue transglutaminase subtype 6 was
Multiple studies have described shared domains be- demonstrated in the cerebellum of patients with celiac
tween rheumatoid arthritis and celiac disease.44 Thus, disease ataxia. Nutritional factors and positive

Nutrition ReviewsV Vol. 75(12):1046–1058


R
1053
anti–transglutaminase 6 autoantibodies might explain Autoimmune thyroiditis
the pathogenesis of the gut–brain axis.121
In an Italian multicenter study, thyroid function was
studied in 241 consecutive untreated patients with
Psoriasis celiac disease and 212 controls. Thyroid disease was 3-
fold higher in patients with celiac disease than in con-
Psoriasis and celiac disease share genetic and inflamma- trols. In most patients who adhered to gluten-free diet
tory features. The connection between psoriasis and ce- for 1 year, a normalization of subclinical hypothyroid-
liac disease was examined in a review of studies of ism was noticed,133 suggesting that, in some patients, a
gluten-free diets in patients with psoriasis.122 Of 33 gluten-free diet may, by itself, reverse abnormal thyroid
patients with psoriasis who were positive for antigliadin levels. Recently, however, an additional study could not
antibodies and were treated with a gluten-free diet, 24 repeat those results.134 In thyroid-associated orbitop-

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


showed a reduction in their psoriatic severity index athy, celiac disease is the only autoimmune disease in
when compared with patients who had psoriasis and which symptoms and tissue transglutaminase levels
were negative for antigliadin antibodies.123 In another usually improve if patients adhere to a gluten-free
28 patients, gluten withdrawal was shown to reduce the diet.135 In a study of the role of gluten in the induction
expression of tissue transglutaminase in patients with of antiendocrine autoantibodies and organ dysfunction
psoriasis who were positive for antigliadin antibod- in adolescent patients with celiac disease, at least 1 anti-
ies.124 Several additional case reports were published in body was positive in 10 of 19 untreated patients but in
which patients with psoriasis experience rapid lesion only 5 of 25 patients treated with gluten-free diets.136 A
resolution following adherence to a gluten-free recent review described the similarities and differences
diet.125–127 There is limited evidence to suggest that between celiac disease and Hashimoto thyroiditis as
gluten restriction may benefit some patients with pso- well as the potential gut–thyroid interrelated path-
riasis, but additional controlled studies in defined ways.137 The concept of a gut–thyroid relationship was
patient populations are needed. supported by the fact that these 2 autoimmune diseases
are related and that tissue transglutaminase antibodies
were described to bind to thyroid follicles and to the ex-
Autoimmune hepatitis and celiac hepatitis tracellular matrix in patients with celiac disease.
Furthermore, anti–tissue transglutaminase titers corre-
Forty percent of individuals have abnormal liver tests late with anti–thyroid peroxidase antibody titers. These
upon the diagnosis of celiac disease, but tests will nor- findings suggest that celiac-associated autoantibodies
malize in the majority who adhere to a gluten-free could contribute to the progression of thyroid dysfunc-
diet.128 Celiac disease is twice as common in patients tion in celiac disease, thus establishing some logical ba-
with cirrhosis as in the normal population, and a sis for the beneficial effects of gluten withdrawal in
gluten-free diet is known to improve liver function patients with autoimmune thyroid disease.137
tests. Abnormally high levels of antiendomysial anti-
bodies and high levels of tissue transglutaminase raise Immunoglobulin A nephropathy
suspicion of celiac disease in patients with cirrhosis and
may prompt physicians to recommend a gluten-free Like other autoimmune diseases, IgA nephropathy
diet.129 Several hepatic and biliary diseases are associ- shares multiple characteristics with celiac disease,138
ated with celiac disease: autoimmune hepatitis, primary and several animal and human studies documented a
sclerosing cholangitis, and primary biliary cirrhosis. beneficial effect of gluten-free diets in this disease.139,140
These liver diseases are less prevalent than celiac hepati-
tis and are associated with increased morbidity and DISCUSSION
mortality, which requires they be treated with appropri-
ate immunosuppressive therapy, rather than just This is the first comprehensive review to address the ad-
gluten-free diets.130 However, a significantly increased verse effects of gluten application or ingestion, in vitro
proportion of patients with coexisting autoimmune or in vivo, and the effect of gluten-free diets in multiple
hepatitis and celiac disease show sustained remission autoimmune diseases: T1D, rheumatoid arthritis, multi-
when compared with patients with autoimmune ple sclerosis, psoriasis, autoimmune hepatitis, and auto-
hepatitis without celiac disease. The improvement in immune thyroiditis. Basic science and translational and
liver function following dietary intervention suggests a clinical research were reviewed to clarify the adverse
possible long-term beneficial effect of a gluten-free effects of gluten and the mechanisms underlying the
diet.131,132 beneficial effects of gluten withdrawal. Summarizing

1054 Nutrition ReviewsV Vol. 75(12):1046–1058


R
Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023
Figure 2 Adverse effects of gluten on the human cellular, immune, intestinal, and systemic compartments. Abbreviations and symbols:
NKG2D, natural killer group 2D costimulatory molecule; PTMP, post-translational modification of proteins; TH-17, T-helper 17 lymphocytes;
TLR4, Toll-like receptor 4 signaling pathway; Treg, regulatory T cells; 䉱, increased; 䉲, decreased.

the evidence from studies that explore the adverse small number of randomized clinical studies and animal
effects of gluten in vitro or of uptake in vivo can help studies, or because studies may have included patients
guide the decision to recommend restriction of dietary with high heterogeneity and varying medical pheno-
gluten to improve the well-being or clinical manage- types, disease activities, and morbidity. There is always
ment of patients with nonceliac chronic conditions. the possibility that some studies were not captured by
However, it should be stressed that the lack of studies in the keywords used in the literature search. Since the
normal human populations devoid of any inflammatory search screened for autoimmune diseases, and not non-
or autoimmune condition limits any recommendation celiac gluten sensitivity, some information could have
to attenuate or prevent gluten consumption worldwide. been missed. Furthermore, the gluten used in the stud-
Since wheat is the most important staple crop in ies might have been contaminated with other soluble
the West, and wheat consumption in countries un- proteins, such as amylase-trypsin inhibitors or yeasts
dergoing urbanization, industrialization, and used in commercial bakeries, which could have affected
Westernization is steadily increasing,3,4,15,63 further the results. Finally, evidence of the adverse effects of
investigations are needed to clarify the drawbacks and gluten was generated mainly in in vitro cell lines and
benefits of gluten ingestion. The enigma is further rein- animal models. There are not yet enough in vivo animal
forced by the gains attributed to gluten, being a major studies or human studies to support a recommendation
source for starch, energy, proteins, vitamins, fiber, and of gluten withdrawal in clinical practice.
phytochemicals. It should be clearly stated that the
above-described response to gluten-free diets in noncel- CONCLUSION
iac autoimmune diseases does not replace the corre-
sponding disease-specific therapies of these disease. The Wheat, along with its main storage protein, gluten, is a
mechanisms of gluten-free diets as nutritional therapy staple nutrient. Increasing evidence shows that gluten
are far from being elucidated, but this review sheds new may have multiple detrimental effects on human health.
light that might clarify some of the pathways underlying Figure 2 summarizes the possible detrimental effects of
the reduction in adverse effects sometimes observed gluten on the intestinal luminal compartment, on the
after gluten withdrawal. immunological pathways, and on cellular as well as
There are, however, some limitations to this review. systemic domains. A word of caution is needed before
The quality of evidence may be limited because of the conclusions are drawn and recommendations are

Nutrition ReviewsV Vol. 75(12):1046–1058


R
1055
suggested. Evidence of the detrimental effects of gluten REFERENCES
is limited to several in vitro and animal studies. Well-
1. Vader LW, Stepniak DT, Bunnik EM, et al. Characterization of cereal toxicity for ce-
designed, controlled studies in humans are lacking. liac disease patients based on protein homology in grains. Gastroenterology.
Thus, no practical clinical recommendations can be 2003;125:1105–1113.
2. Food and Agriculture Organization of the United Nations: FAO cereal supply and
suggested until further clinical trials are performed. demand brief: summary tables (world wheat market). http://www.fao.org/world
Implementing gluten-free diets in certain individuals af- foodsituation/csdb/en/. Updated October 5, 2017. Accessed October 5, 2017.
3. Lerner A, Jeremias P, Matthias T. The world incidence and prevalence of autoim-
fected by nonceliac autoimmune disease might be in- mune diseases is increasing: Na review. Int J Celiac Dis. 2016;3:151–155.
strumental in minimizing the adverse effects of gluten 4. Lerner A, Jermias P, Matthias T. The world incidence of celiac disease is increas-
ing: a review. Int J Recent Sci Res. 2015;7:5491–5496.
in humans, but for now, the use of such nutritional 5. Lebwohl B, Ludvigsson JF, Green PHR. The unfolding story of celiac disease risk
therapy is not based on adequate human investigations. factors. Clin Gastroenterol Hepatol. 2014;12:632–635.
6. Tucci F, Astarita L, Abkari A, et al. Celiac disease in the Mediterranean area. BMC
Considering the effects of gluten on the microbiome, Gastroenterol. 2014;14:24.
the role of gluten in oxidative stress and epigenetics, 7. Kang JY, Kang AH, Green A, et al. Systematic review: worldwide variation in the

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


frequency of coeliac disease and changes over time. Aliment Pharmacol Ther.
and the relationship of gluten to the induction and pro- 2013;38:226–245.
gression of autoimmunity, a gluten-free diet might have 8. Guandalini S, Polanco I. Nonceliac gluten sensitivity or wheat intolerance syn-
drome? J Pediatr. 2015;166:805–811.
multiple indirect consequences.42,63,141–143 9. Tovoli F, Masi C, Guidetti E, et al. Clinical and diagnostic aspects of gluten related
Currently, there is no clear indication to withdraw disorders. World J Clin Cases. 2015;3:275–284.
10. Volta U, Caio G, Tovoli F, et al. Non-celiac gluten sensitivity: questions still to be
gluten in non–gluten-related conditions. However, answered despite increasing awareness. Cell Mol Immunol. 2013;10:383–392.
since there is an increased prevalence of celiac disease 11. Belderok B. Developments in bread-making processes. Plant Foods Hum Nutr.
in multiple autoimmune diseases and vice versa, it is 2000;55:1–86.
12. Gobbetti M, Giuseppe Rizzello C, Di Cagno R, et al. Sourdough lactobacilli and ce-
suggested that patients with autoimmune disease be liac disease. Food Microbiol. 2007;24:187–196.
screened for antibiodies associated with celiac disease. 13. Catassi C, Kryszak D, Bhatti B, et al. Natural history of celiac disease autoimmunity
in a USA cohort followed since 1974. Ann Med. 2010;42:530–538.
In view of the central role played by the human 14. Catassi C, Fasano A. Tempters and gluten-free diet. Nutrients. 2016;8:786–787.
intestine in inducing autoimmunity in peripheral 15. Catassi C, Gatti S, Lionetti E. World perspective and celiac disease epidemiology.
Dig Dis. 2015;33:141–146.
organs,44,98,99,137,138,144,145 there is a need to elucidate the 16. Larsen J, Weile C, Antvorskov JC, et al. Effect of dietary gluten on dendritic cells and
pathways, the molecules, or the messengers involved in innate immune subsets in BALB/c and NOD mice. PLoS One. 2015;10:e0118618.
17. Larsen J, Dall M, Antvorskov JC, et al. Dietary gluten increases natural killer cell
the interplay between gluten and nonceliac autoimmune cytotoxicity and cytokine secretion. Eur J Immunol. 2014;44:3056–3067.
conditions. It is hoped that the detrimental effects of glu- 18. Antvorskov JC, Fundova P, Buschard K, et al. Impact of dietary gluten on regula-
tory T cells and Th17 cells in BALB/c mice. PLoS One. 2012;7:e33315. doi:10.
ten summarized here will encourage the clinical and the 1371/journal.pone.0033315
scientific communities to explore the underlying mecha- 19. Dall M, Calloe K, Haupt-Jorgensen M, et al. Gliadin fragments and a specific glia-
din 33-mer peptide close KATP channels and induce insulin secretion in INS-1E
nistic pathways and verify whether observations in ani- cells and rat islets of Langerhans. PLoS One. 2013;8:e66474.
mals and ex vivo can be substantiated in humans. 20. Lammers KM, Chieppa M, Liu L, et al. Gliadin induces neutrophil migration via
engagement of the formyl peptide receptor, FPR1. PLoS One. 2015;10:e0138338.
doi: 10.1371/journal.pone.0138338
Acknowledgments 21. Adlercreutz EH, Weile C, Larsen J, et al. A gluten-free diet lowers NKG2D and li-
gand expression in BALB/c and non-obese diabetic (NOD) mice. Clin Exp
Immunol. 2014;177:391–403.
The authors would like to acknowledge Mr Neu Alf, 22. Junker Y, Zeissig S, Kim SJ, et al. Wheat amylase trypsin inhibitors drive intestinal in-
medical illustrator, for providing the figures and flammation via activation of toll-like receptor 4. J Exp Med. 2012;209:2395–2408.
23. Myles IA. Fast food fever: reviewing the impacts of the Western diet on immu-
Dr. Ramesh Ajay for editing the manuscript. nity. Nutr J. 2014;13:61–77.
24. Thomas KE, Sapone A, Fasano A, et al. Gliadin stimulation of murine macrophage
Author contributions A.L. wrote the manuscript and inflammatory gene expression and intestinal permeability are MyD88-
dependent: role of the innate immune response in celiac disease. J Immunol.
was responsible for the study concept, the study design, 2006;176:2512–2521.
and the literature review. Y.S. supervised and was re- 25. Tuckova L, Flegelova Z, Tlaskalova-Hogenova H, et al. Activation of macrophages
sponsible for the study concept, the interpretation of by food antigens: enhancing effect of gluten on nitric oxide and cytokine pro-
duction. J Leukoc Biol. 2000;67:312–318.
data, and the editing and critical revision of the manu- 26. Tuckova L, Novotna J, Novak P, et al. Activation of macrophages by gliadin frag-
script. T.M. was responsible for the study design and ments: isolation and characterization of active peptide. J Leukoc Biol.
2002;71:625–631.
the critical revision of the manuscript and provided ad- 27. Nikulina M, Habich C, Flohe SB, et al. Wheat gluten causes dendritic cell matura-
ministrative, technical, and material support. All tion and chemokine secretion. J Immunol. 2004;173:1925–1933.
28. Palova-Jelinkova L, Rozkova D, Pecharova B, et al. Gliadin fragments induce phe-
authors approved the final version of the manuscript, notypic and functional maturation of human dendritic cells. J Immunol.
including the list of authors. 2005;175:7038–7045.
29. Ciccocioppo R, Rossi M, Pesce I, et al. Effects of gliadin stimulation on bone
marrow-derived dendritic cells from HLA-DQ8 transgenic MICE. Digt Liver Dis.
Funding/support. No external funds supported this 2008;40:927–935.
30. Gujral N, Suh JW, Sunwoo HH. Effect of anti-gliadin IgY antibody on epithelial in-
work. testinal integrity and inflammatory response induced by gliadin. BMC Immunol.
2015;16:41–51.
31. Flohe SB, Wasmuth HE, Kerad JB, et al. A wheat-based, diabetes-promoting diet
Declaration of interest. The authors have no relevant induces a Th1-type cytokine bias in the gut of NOD mice. Cytokine.
interests to declare. 2003;21:149–154.

1056 Nutrition ReviewsV Vol. 75(12):1046–1058


R
32. Alam C, Valkonen S, Palagani V, et al. Inflammatory tendencies and overproduc- 58. Dolfini E, Elli L, Ferrero S, et al. Bread wheat gliadin cytotoxicity: a new three-
tion of IL-17 in the colon of young NOD mice are counteracted with diet change. dimensional cell model. Scand J Clin Lab Invest. 2003;63:135–141.
Diabetes. 2010;59:2237–2246. 59. Dolfini E, Elli L, Roncoroni L, et al. Damaging effects of gliadin on three-
33. Scott FW, Rowsell P, Wang GS, et al. Oral exposure to diabetes-promoting food dimensional cell culture model. World J Gastroenterol.
or immunomodulators in neonates alters gut cytokines and diabetes. Diabetes. 2005;11:5973–5977.
2002;51:73–78. 60. Giovannini C, Matarrese P, Scazzocchio B, et al. Wheat gliadin induces apoptosis
34. Scott FW, Cloutier HE, Kleemann R, et al. Potential mechanisms by which certain of intestinal cells via an autocrine mechanism involving Fas-Fas ligand pathway.
foods promote or inhibit the development of spontaneous diabetes in BB rats: FEBS Lett. 2003;540:117–124.
dose, timing, early effect on islet area, and switch in infiltrate from Th1 to Th2 61. Giovannini C, Sanchez M, Straface E, et al. Induction of apoptosis in Caco-2 cells
cells. Diabetes. 1997;46:589–598. by wheat gliadin peptides. Toxicology. 2000;145:63–71.
35. Antvorskov JC, Fundova P, Buschard K, et al. Dietary gluten alters the balance of 62. Hollon J, Puppa EL, Greenwald B, et al. Effect of gliadin on permeability of intesti-
pro-inflammatory and anti-inflammatory cytokines in T cells of BALB/c mice. nal biopsy explants from celiac disease patients and patients with non-celiac glu-
Immunology. 2013;138:23–33. ten sensitivity. Nutrients. 2015;7:1565–1576.
36. Bernardo D, Garrote JA, Fernandez-Salazar L, et al. Is gliadin really safe for 63. Lerner A, Matthias T. Changes in intestinal tight junction permeability associated
non-coeliac individuals? Production of interleukin 15 in biopsy culture with industrial food additives explain the rising incidence of autoimmune dis-
from non-coeliac individuals challenged with gliadin peptides. Gut. ease. Autoimm Rev. 2015;14:479–489.
2007;56:889–890. 64. Clemente MG, De Virgiliis S, Kang JS, et al. Early effects of gliadin on enterocyte

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


37. Palova-Jelinkova L, Da nova K, Drasarova H, et al. Pepsin digest of wheat gliadin intracellular signalling involved in intestinal barrier function. Gut.
fraction increases production of IL-1b via TLR4/MyD88/TRIF/MAPK/NF-jB signal- 2003;52:218–223.
ing pathway and an NLRP3 inflammasome activation. PLoS One. 2013;8:e62426. 65. De Santis S, Cavalcanti E, Mastronardi M, et al. Nutritional keys for intestinal bar-
doi:10.1371/journal.pone.0062426 rier modulation. Front Immunol. 2015;6:612–626.
38. Stepankova R, Tlaskalova-Hogenova H, Sinkora J, et al. Changes in jejunal mu- 66. Zimmermann C, Rudloff S, Lochnit G, et al. Epithelial transport of immunogenic
cosa after long-term feeding of germfree rats with gluten. Scand J Gastroenterol. and toxic gliadin peptides in vitro. PLoS One. 2014;9:e113932. doi:10.1371/jour
1996;31:551–557. nal.pone.0113932
39. Skovbjerg H, Norén O, Anthonsen D, et al. Gliadin is a good substrate of several 67. Antvorskov JC, Josefsen K, Engkilde K, et al. Dietary gluten and the development
transglutaminases: possible implication in the pathogenesis of celiac disease. of type 1 diabetes. Diabetologia. 2014;57:1770–1780.
Scand J Gastroenterol. 2002;37:812–817. 68. Hansen AK, Ling F, Kaas A, et al. Diabetes preventive gluten-free diet decreases
40. Lerner A, Matthias T. Possible association between celiac disease and bacterial the number of caecal bacteria in non-obese diabetic mice. Diabetes Metab Res
transglutaminase in food processing: a hypothesis. Nutr Rev. 2015;73:544–552. Rev. 2006;22:220–225.
41. Lerner A, Matthias T. Food industrial microbial transglutaminase in celiac disease: 69. Marietta EV, Gomez AM, Yeoman C, et al. Low incidence of spontaneous type 1
treat or trick. Int J Celiac Dis. 2015;3:1–6. diabetes in non-obese diabetic mice raised on gluten-free diets is associated
42. Lerner A, Trinder P, Matthias T. Breaching intestinal tight junction permeability with changes in the intestinal microbiome. PLoS One. 2013;8:e78687. doi:10.
associated with industrial food additives might explain the surge in autoimmune 1371/journal.pone.0078687
disease incidence. In: Conrad K, Chan EKL, Andrade LEC, eds. Autoantigens, 70. Sanz Y. Microbiome and gluten. Ann Nutr Metab. 2015;67(suppl 2):28–41.
Autoantibodies, Autoimmunity. Vol. 10. Lengerich, Germany: Pabst Science 71. Carding S, Verbeke K, Vipond DT, et al. Dysbiosis of the gut microbiota in disease.
Publishers; 2015:10–16. Microb Ecol Health Dis. 2015;26:26191. doi:10.3402/mehd.v26.26191 [
43. Lerner A, Neidhofer S, Matthias T. Transglutaminase 2 and anti transglutaminase 72. Mohan M, Chow CT, Ryan CN, et al. Dietary gluten-induced gut dysbiosis is ac-
2 autoantibodies in celiac disease and beyond: part A: TG2 double-edged sword: companied by selective upregulation of microRNAs with intestinal tight junction
gut and extraintestinal involvement. Immunome Res. 2015;11:3. doi:10.4172/ and bacteria-binding motifs in rhesus macaque model of celiac disease.
1745-7580.10000101 Nutrients. 2016;8:684–701.
44. Lerner A, Matthias T. Rheumatoid arthritis–celiac disease relationship: joints get 73. Marasco G, Di Biase AR, Schiumerini R, et al. Gut microbiota and celiac disease.
that gut feeling. Autoimm Rev. 2015;14:1038–1047. Dig Dis Sci. 2016;61:1461–1472.
45. Lerner A, Aminov R, Matthias T. Dysbiosis may trigger autoimmune diseases via 74. Bonder MJ, Tigchelaar EF, Cai X, et al. The influence of a short-term gluten-free
inappropriate posttranslational modification of host proteins. Front Microbiol. diet on the human gut microbiome. Genome Med. 2016;8:45. doi:10.1186/
2016;7:84–89. s13073-016-0295-y
46. Lerner A, Aminov R, Matthias T. Intestinal dysbiotic transglutaminases are poten- 75. De Angelis M, Vannini L, Di Cagno R, et al. Salivary and fecal microbiota and
tial environmental drivers of systemic autoimmunogenesis. Front Microbiol. metabolome of celiac children under gluten-free diet. Int J Food Microbiol.
2017;8:66. doi:10.3389/fmicb.2017.00066 2016;239:125–132.
47. Kumar V, Beutner EH, Lerner A, et al. Comparison of disease specificity of antien- 76. Caminero A, Galipeau HJ, McCarville JL, et al. Duodenal bacteria from patients
domysial and antigliadin antibodies. In: Beutner EH, Chorzelski TP, Kumar V, eds. with celiac disease and healthy subjects distinctly affect gluten breakdown and
Immunopathology of the Skin. New York, NY: John Wiley; 1987: 483–488. immunogenicity. Gastroenterology. 2016;151:670–683.
48. Tucker NT, Barghuthy FS, Prihoda TJ, et al. Antigliadin antibodies detected by 77. Hudson DA, Purdham DR, Cornell HJ, et al. Nonspecific cytotoxicity of wheat glia-
enzyme-linked immunosorbent assay (ELISA) as a marker of childhood celiac dis- din components towards cultured human cells. Lancet. 1976;307:339–341.
ease. J Pediatr. 1988;113:286–289. 78. Ferretti G, Bacchetti T, Masciangelo S, et al. Celiac disease, inflammation and oxi-
49. Lerner A, Kumar V, Iancu TC. Immunological diagnosis of childhood celiac dis- dative damage: a nutrigenetic approach. Nutrients. 2012;4:243–257.
ease: comparison between antigliadin, antireticulin and antiendomysial antibod- 79. Trivedi MS, Shah JS, Al-Mughairy S, et al. Food-derived opioid peptides inhibit
ies. Clin Exp Immunol. 1994;95:78–82. cysteine uptake with redox and epigenetic consequences. J Nutr Biochem.
50. Lerner A, Lebenthal E. The controversy of antigluten antibody (AGA) as a diag- 2014;25:1011–1018.
nostic tool in celiac disease. J Pediatr Gastroenterol Nutr. 1991;12:407–409. 80. Luciani A, Villella VR, Vasaturo A, et al. Lysosomal accumulation of gliadin p31-43
51. Vriezinga SL, Auricchio R, Bravi E, et al. Randomized feeding intervention in peptide induces oxidative stress and tissue transglutaminase-mediated
infants at high risk for celiac disease. N Engl J Med. 2014;371:1304–1315. PPARgamma downregulation in intestinal epithelial cells and coeliac mucosa.
52. Hvatum M, Kanerud L, Hallgren R, et al. The gut–joint axis: cross reactive food Gut. 2010;59:311–319.
antibodies in rheumatoid arthritis. Gut. 2006;55:1240–1247. 81. Serena G, Yan S, Camhi S, et al. Proinflammatory cytokine interferon-c and
53. Vojdani A. Detection of IgE, IgG, IgA and IgM antibodies against raw and proc- microbiome-derived metabolites dictate epigenetic switch between forkhead
essed food antigens. Nutr Metab (London). 2009;6:22–39. box protein 3 isoforms in coeliac disease. Clin Exp Immunol. 2017;187:490–506.
54. Kaufmann A, Koppel R, Widmer M. Determination of microbial transglutaminase 82. Giovannini C, Maiuri L, De Vincenzi M. Cytotoxic effect of prolamin-derived pepti-
in meat and meat products. Food Addit Contam Part A Chem Anal Control Expo des on in vitro cultures of cell line Caco-2: implications for coeliac disease.
Risk Assess. 2012;29:1364–1373. Toxicol in Vitro. 1995;9:251–255.
55. Lerner A, Neidhofer S, Jeremias P, et al. The diversities between the neo-epitope 83. Giovannini C, Mancini E, De Vincenzi M. Inhibition of the cellular metabolism of
and the IgA-tissue transglutaminase autoantibodies in celiac disease. In: Conrad Caco-2 cells by prolamin peptides from cereals toxic for coeliacs. Toxicol in Vitro.
K, Chan EKL, Andrade LEC, eds. Autoantigens, Autoantibodies, Autoimmunity. 1996;10:533–538.
Vol. 10. Lengerich, Germany: Pabst Science Publishers; 2015:220–226. 84. Elli L, Dolfini E, Bardella MT. Gliadin cytotoxicity and in vitro cell cultures. Toxicol
56. Lerner A, Jeremias P, Neidhofer S, et al. Antibodies against neo-epitope tTg com- Lett. 2003;146:1–8.
plexed to gliadin are different and more reliable then anti-tTg for the diagnosis 85. Bressan P, Kramer P. Bread and other edible agents of mental disease. Front
of pediatric celiac disease. J Immunol Meth. 2016;429:15–20. Hum Neurosci. 2016;10:130–140.
57. Matthias T, Jeremias P, Neidhofer S, et al. The industrial food additive microbial 86. Karakuła-Juchnowicz H, Dzikowski M, Pelczarska A, et al. The brain-gut axis dys-
transglutaminase, mimics the tissue transglutaminase and is immunogenic in ce- functions and hypersensitivity to food antigens in the etiopathogenesis of
liac disease patients. Autoimm Rev. 2016;15:111–1119. schizophrenia. Psychiatr Pol. 2016;50:747–760.

Nutrition ReviewsV Vol. 75(12):1046–1058


R
1057
87. Ghalichi F, Ghaemmaghami J, Malek A, et al. Effect of gluten free diet on gastro- 116. Elkan AC, Sjoberg B, Kolsrud B, et al. Gluten-free vegan diet induces decreased
intestinal and behavioral indices for children with autism spectrum disorders: a LDL and oxidized LDL levels and raised atheroprotective natural antibodies
randomized clinical trial. World J Pediatr. 2016;12:436–442. against phosphorylcholine in patients with rheumatoid arthritis: a randomized
88. Nemani K, Hosseini Ghomi R, McCormick B, et al. Schizophrenia and the gut– study. Arthritis Res Ther. 2008;10:R34–R41.
brain axis. Prog Neuropsychopharmacol Biol Psychiatry. 2015;56:155–160. 117. Shor DB, Barzilai O, Ram M, et al. Gluten sensitivity in multiple sclerosis: experi-
89. Lionetti E, Leonardi S, Franzonello C, et al. Gluten psychosis: confirmation of a mental myth or clinical truth? Ann N Y Acad Sci. 2009;1173:343–349.
new clinical entity. Nutrients. 2015;7:5532–5539. 118. Rodrigo L, Hernandez-Lahoz C, Fuentes D, et al. Prevalence of celiac disease in
90. Choi S, Disilvio B, Fernstrom MH, et al. Meal ingestion, amino acids and brain multiple sclerosis. BMC Neurol. 2011;11:31–37.
neurotransmitters: effects of dietary protein source on serotonin and catechol- 119. Finsterer J, Leutmezer F. Celiac disease with cerebral and peripheral nerve in-
amine synthesis rates. Physiol Behav. 2009;98:156–162. volvement mimicking multiple sclerosis. J Med Life. 2014;7:440–444.
91. Casella S, Zanini B, Lanzarotto F, et al. Cognitive performance is impaired in coe- 120. von Geldern G, Mowry EM. The influence of nutritional factors on the prognosis
liac patients on gluten free diet: a case–control study in patients older than 65 of multiple sclerosis. Nat Rev Neurol. 2012;8:678–689.
years of age. Dig Liver Dis. 2012;44:729–735. 121. McKeon A, Lennon VA, Pittock SJ, et al. The neurologic significance of celiac dis-
92. Lerner A, Matthias T. Increased knowledge and awareness of celiac disease will ease biomarkers. Neurology. 2014;83:1789–1796.
benefit the elderly. Int J Celiac Dis. 2016;3:112–114. 122. Bhatia BK, Millsop JW, Debbaneh M, et al. Diet and psoriasis, part II: celiac disease
93. Zelnik N, Pacht A, Obeid R, et al. Range of neurological disorders in patients with and role of a gluten-free diet. J Am Acad Dermatol. 2014;71:350–358.
celiac disease. Pediatrics. 2004;113:1672–1676. 123. Michaelsson G, Gerden B, Hagforsen E, et al. Psoriasis patients with antibodies to

Downloaded from https://academic.oup.com/nutritionreviews/article/75/12/1046/4675264 by guest on 26 July 2023


94. Lerner A, Makhoul BF, Eliakim R. Neurological manifestations of celiac disease in gliadin can be improved by a gluten-free diet. Br J Dermatol. 2000;142:44–51.
children and adults. Eur Neurol J. 2012;4:15–20. 124. Michaelsson G, Ahs S, Hammarstrom I, et al. Gluten-free diet in psoriasis patients
95. Parisi P, Pietropaoli N, Ferretti A, et al. Role of the gluten-free diet on with antibodies to gliadin results in decreased expression of tissue transglutami-
neurological-EEG findings and sleep disordered breathing in children with celiac nase and fewer Ki67þ cells in the dermis. Acta Derm Venereol.
disease. Seizure. 2015;25:181–183. 2003;83:425–429.
96. Bella R, Lanza G, Cantone M, et al. Effect of a gluten-free diet on cortical excitabil- 125. Addolorato G, Parente A, de Lorenzi G, et al. Rapid regression of psoriasis in a
ity in adults with celiac disease. PLoS One. 2015;10:e0129218. coeliac patient after gluten-free diet. A case report and review of the literature.
97. Fanciulli G, Dettori A, Demontis MP, et al. Gluten exorphin B5 stimulates prolac- Digestion. 2003;68:9–12.
tin secretion through opioid receptors located outside the blood-brain barrier. 126. Frikha F, Snoussi M, Bahloul Z. Osteomalacia associated with cutaneous psoriasis
Life Sci. 2005;76:1713–1719. as the presenting feature of coeliac disease: a case report. Pan Afr Med J.
98. Lerner A, Matthias T. GUT-the Trojan horse in remote organs’ autoimmunity. J 2012;11:58. doi:10.11604/pamj.2012.11.58.1613
Clin Cell Immunol. 2016;7:401–410. 127. De Boer WA, Tytgat GN. A patient with osteomalacia as single presenting symp-
99. Koning F. Adverse effects of wheat gluten. Ann Nutr Metab. 2015;67:8–14. tom of gluten-sensitive enteropathy. J Intern Med. 1992;232:81–85.
100. Zheng J, Petersen F, Yu X. The role of PTPN22 in autoimmunity: learning from 128. Castillo NE, Vanga RR, Theethira TG, et al. Prevalence of abnormal liver
mice. Autoimmun Rev. 2014;13:266–271. function tests in celiac disease and the effect of a gluten-free diet in the US pop-
101. Richard-Miceli C, Criswell LA. Emerging patterns of genetic overlap across auto- ulation. Am J Gastroenterol. 2015;110:1216–1222.
immune disorders. Genome Med. 2012;4:6–14. 129. Wakim-Fleming J, Pagadala MR, McCullough AJ, et al. Prevalence of celiac dis-
102. Gutierrez-Achury J, Coutinho de Almeida R, Wijmenga C. Shared genetics in coe- ease in cirrhosis and outcome of cirrhosis on a gluten free diet: a prospective
liac disease and other immune-mediated diseases. J Intern Med. study. J Hepatol. 2014;61:558–563.
2011;269:591–603. 130. Anania C, De Luca E, De Castro G, et al. Liver involvement in pediatric celiac dis-
103. Zhernakova A, Stahl EA, Trynka G, et al. Meta-analysis of genome-wide associa- ease. World J Gastroenterol. 2015;21:5813–5822.
tion studies in celiac disease and rheumatoid arthritis identifies fourteen non- 131. Nastasio S, Sciveres M, Riva S, et al. Celiac disease-associated autoimmune hepa-
HLA shared loci. PLoS Genet. 2011;7:e1002004. doi:10.1371/journal.pgen. titis in childhood: long-term response to treatment. J Pediatr Gastroenterol Nutr.
1002004 2013;56:671–674.
104. Fortune MD, Guo H, Burren O, et al. Statistical colocalization of genetic risk var- 132. Colecchia A, Di Biase AR, Scaioli E, et al. Coeliac disease and autoimmune hepati-
iants for related autoimmune diseases in the context of common controls. Nat tis: gluten-free diet can influence liver disease outcome. Dig Liv Dis.
Genet. 2015;47:839–846. 2011;43:247–249.
105. Cosnes J, Cellier C, Viola S, et al. Incidence of autoimmune diseases in celiac dis- 133. Sategna-Guidetti C, Volta U, Ciacci C, et al. Prevalence of thyroid disorders in
ease: protective effect of the gluten-free diet. Clin Gastroenterol Hepatol. untreated adult celiac disease patients and effect of gluten withdrawal: an
2008;6:753–758. Italian multicenter study. Am J Gastroenterol. 2001;96:751–757.
106. San Mauro Martin I, Garicano Vilar E, Collado Yurrutia L, et al. Is gluten the great 134. Metso S, Hyytia-Ilmonen H, Kaukinen K, et al. Gluten-free diet and autoimmune
etiopathogenic agent of disease in the XXI century [in Spanish]?. Nutr Hosp. thyroiditis in patients with celiac disease. A prospective controlled study. Scand J
2014;30:1203–1210. Gastroenterol. 2012;47:43–48.
107. El-Chammas K, Danner E. Gluten-free diet in nonceliac disease. Nutr Clin Pract. 135. Ponto KA, Schuppan D, Zwiener I, et al. Thyroid-associated orbitopathy is linked
2011;26:294–299. to gastrointestinal autoimmunity. Clin Exp Immunol. 2014;178:57–64.
108. MacFarlane AJ, Burghardt KM, Kelly J, et al. A type 1 diabetes-related protein 136. Toscano V, Conti FG, Anastasi E, et al. Importance of gluten in the induction of
from wheat (Triticum aestivum): cDNA clone of a wheat storage globulin, Glb1, endocrine autoantibodies and organ dysfunction in adolescent celiac patients.
linked to islet damage. J Biol Chem. 2003;278:54–63. Am J Gastroenterol. 2000;95:1742–1748.
109. Loit E, Melnyk CW, MacFarlane AJ, et al. Identification of three wheat globulin 137. Lerner A, Jeremias P, Matthias T. The gut-thyroid axis and celiac disease. Endocr
genes by screening a Triticum aestivum BAC genomic library with cDNA from a Connect. 2017;6:R52–R58.
diabetes-associated globulin. BMC Plant Biol. 2009;9:93–103. 138. Lerner A, Berthelot L, Jeremias P, et al. Gluten, transglutaminase, celiac disease
110. Galipeau HJ, Rulli NE, Jury J, et al. Sensitization to gliadin induces moderate en- and IgA nephropathy. J Clin Cell Immunol. 2017;8:499–503.
teropathy and insulitis in nonobese diabetic-DQ8 mice. J Immunol. 139. Papista C, Lechner S, Ben Mkaddem S, et al. Gluten exacerbates IgA nephropathy
2011;187:4338–4346. in humanized mice through gliadin–CD89 interaction. Kidney Int.
111. Funda DP, Kaas A, Bock T, et al. Gluten-free diet prevents diabetes in NOD mice. 2015;88:276–285.
Diabetes Metab Res Rev. 1999;15:323–327. 140. Koivuviita N, Tertti R, Heiro M, et al. A case report: a patient with IgA nephropa-
112. Pastore MR, Bazzigaluppi E, Belloni C, et al. Six months of gluten-free diet do not thy and coeliac disease. Complete clinical remission following gluten-free diet.
influence autoantibody titers, but improve insulin secretion in subjects at high NDT Plus. 2009;2:161–163.
risk for type 1 diabetes. J Clin Endocrinol Metabol. 2003;88:162–165. 141. Shamriz O, Mizrahi H, Werbner M, et al. Microbiota at the crossroads of autoim-
113. Hummel M, Bonifacio E, Naserke HE, et al. Elimination of dietary gluten does munity. Autoimmun Rev. 2016;15:859–869.
not reduce titers of type 1 diabetes-associated autoantibodies in high-risk sub- 142. Ortona E, Maselli A, Delunardo F, et al. Relationship between redox status and
jects. Diabetes Care. 2002;25:1111–1116. cell fate in immunity and autoimmunity. Antioxid Redox Signal.
114. Fuchtenbusch M, Ziegler AG, Hummel M. Elimination of dietary gluten and de- 2014;21:103–122.
velopment of type 1 diabetes in high risk subjects. Rev Diabet Stud. 143. Youinou P, Pers JO, Gershwin ME, et al. Geo-epidemiology and autoimmunity.
2004;1:39–41. J Autoimmun. 2010;34:J163–J167.
115. Hafstrom I, Ringertz B, Spangberg A, et al. A vegan diet free of gluten improves 144. Lerner A, Neidhofer S, Matthias T. The gut-gut axis: cohabitation of celiac,
the signs and symptoms of rheumatoid arthritis: the effects on arthritis correlate Crohn’s disease and IgA deficiency. Int J Celiac Dis. 2016;4:68–70.
with a reduction in antibodies to food antigens. Rheumatology (Oxford). 145. Lerner A, Matthias T. Gut-bone cross talks and implications in celiac disease. Int J
2001;40:1175–1179. Celiac Dis. 2016;4:19–23.

1058 Nutrition ReviewsV Vol. 75(12):1046–1058


R

You might also like