Next Article in Journal
Inhibition of Xanthine Oxidase by Four Phenolic Acids: Kinetic, Spectroscopic, Molecular Simulation, and Cellular Insights
Previous Article in Journal
Impact of Tissue Decay on Drying Kinetics, Moisture Diffusivity, and Microstructure of Bell Pepper and Strawberry
Previous Article in Special Issue
Immunomodulatory, Anticancer, and Antioxidative Activities of Bioactive Peptide Fractions from Enzymatically Hydrolyzed White Jellyfish (Lobonema smithii)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gluten Proteins: Beneficial Factors and Toxic Triggers in Human Health

by
Luigia Di Stasio
and
Gianfranco Mamone
*
Institute of Food Sciences, National Research Council, 83100 Avellino, Italy
*
Author to whom correspondence should be addressed.
Foods 2025, 14(19), 3403; https://doi.org/10.3390/foods14193403
Submission received: 31 July 2025 / Revised: 15 September 2025 / Accepted: 27 September 2025 / Published: 1 October 2025

Abstract

The impact of gluten on human health has been the subject of intense study. Gluten proteins are implicated in a range of adverse health effects, such as allergy, celiac disease, and non-celiac gluten sensitivity in predisposed individuals. However, beyond their potential to trigger adverse reactions, gluten proteins also harbor sequences that, upon digestion or fermentation, can release bioactive peptides with health-promoting properties. These peptides have been reported to exhibit antioxidant, antihypertensive, and immunomodulatory activities, suggesting that gluten-derived products may contribute positively to human health. This review aims to explore the dual nature of gluten proteins, examining their role as both potential health hazards and sources of beneficial molecules. By dissecting the molecular mechanisms underlying gluten-related disorders and the functional properties of gluten-derived peptides, we seek to provide a balanced view of gluten’s complex role in nutrition and health.

Graphical Abstract

1. Introduction

Gluten is a complex mixture of storage proteins predominantly found in the endosperm of wheat (Triticum species) and, to a lesser extent, in other cereals such as barley and rye. Because of its unique rheological and functional properties, gluten is a critical component in food processing, particularly in the production of pasta and bakery food products. Gluten also affects consumer acceptability, especially with regard to product texture and appearance. The quantity and quality of gluten proteins are key criteria in wheat breeding and flour quality assessment [1]. Given its central role in food formulation and processing, gluten remains a highly valued ingredient in the global food industry, despite growing health concerns among certain populations, since its consumption is also associated with a spectrum of adverse health effects in susceptible individuals. The most well-characterized condition is celiac disease (CD), an autoimmune enteropathy triggered by the ingestion of gluten peptides in genetically predisposed individuals. CD prevalence in the general population is estimated at approximately 1% [2]. Other gluten-related disorders include non-celiac gluten sensitivity (NCGS), which is characterized by gastrointestinal and extraintestinal symptoms in the absence of celiac-specific serology or histological findings, and wheat allergy, an IgE-mediated hypersensitivity reaction [3].
Despite these well-documented adverse effects, gluten proteins are also a source of peptides with potential health-promoting properties, which are released during the enzymatic hydrolysis process, such as those naturally occurring during gastrointestinal digestion, or during fermentation mediated by microbial proteases [4].
The contrasting roles of gluten, as both a health risk and a potential source of beneficial compounds, highlight the need for a comprehensive and evidence-based evaluation of its effects on human health. The recent ongoing research has focused on peptide identification, functional validation, and controlled food processing strategies, which are helpful to mitigate gluten-related risks while enhancing its positive contributions to nutrition and well-being [5].
In light of the duality that characterizes gluten proteins, being both potential health hazards and sources of beneficial molecules, this review aims to provide a comprehensive overview of their structural, functional, and biological properties. We first describe the molecular composition and classification of gluten proteins, with an emphasis on their unique physicochemical features relevant to food processing. We then explore the mechanisms underlying gluten-related disorders, including CD, NCGS, and wheat allergy, highlighting the specific peptide sequences and immune pathways involved. Finally, we examine emerging evidence on gluten-derived bioactive peptides and their potential health-promoting effects, focusing on their generation during digestion and fermentation.
By integrating insights from food science, immunology, and nutritional biochemistry, this review seeks to present a balanced and updated perspective on gluten, moving beyond its role as a dietary trigger of disease to consider its possible functional applications in human health.

2. Structure and Composition of Gluten Proteins

2.1. Classification and Structure of Gluten Proteins

According to the Osborne classification based on solubility in aqueous alcohols, gluten proteins can be classified into two main fractions: soluble-monomeric gliadins and the insoluble-polymeric glutenins [1]. Furthermore, based on electrophoretic mobility and molecular weights (MWs), gliadins are classified in four different types: ω5, ω1,2, α/β, and γ-gliadins, which have also been redefined based on their amino acid sequences as sulphur (S)-rich (α- and γ-type) and S-poor (ω-type) prolamins [6]. The gliadins ω5- and ω1,2-gliadins are characterized by a high content of glutamine, proline, and phenylalanine (but poor in cysteine), and their repetitive sequences distinguish almost entirely their protein structure (e.g., such as QQPQQPFP in ω1,2 gliadins and QQQFP in ω5-gliadins). While α/β and γ-gliadins are characterized by a much lower content of glutamine and proline than ω-gliadins, and their N-terminal and C-terminal profile are clearly distinct. Within the C-terminal domains, in fact, α/β- and γ-gliadins are almost homologous. Although the N-terminal domain, representing about 50% of the total structure, consists mostly of repetitive sequences rich in glutamine, proline, phenylalanine, and tyrosine (e.g., the dodecapeptides QPQPFPQQPYP, which are usually repeated five times in α/β- gliadins; QPQPFP and PQQPYP in α-gliadins, and the QPQQPFP in γ-gliadins, which is repeated up to 16 times) [7,8].
Glutenins, following the reduction of inter-chain and intra-chain disulfide bonds, consist of two main groups named high-molecular-weight (HMW)-GS and low-molecular-weight (LMW)-GS. LMW-GS can be further subdivided into B, C, and D-type, based on their SDS-PAGE mobility and isoelectric point (pI). While on the basis of N-terminal amino acid sequences, three subgroups of typical LMW-GS can be classified, called LMW-s, LMW-m, and LMW-i types, according to the first amino acid residue of the mature protein: serine, methionine, or isoleucine, respectively. HMW-GS are subdivided into x- and y-types, encoded by closely linked genes at the Glu-A1 and Glu-B1 loci. Structurally, HMW-GS consists of three domains characterized by a C-terminal domain, which contains cysteine residues involved in inter-chain and intra-chain disulfide bonds, a non-repetitive N-terminal domain, and a repetitive central domain that promotes intermolecular hydrogen bonding and that is characterized by repetitive units comprising mainly glutamine, glycine, and proline, such as PGQGQQ (both types), GYYPTSPQQ or GYYPTSLQQ, and GQQ (x-type), and HYPASQ (y-type) [6,7,8,9,10].
The formation of the gluten network during dough mixing involves a complex series of reactions and molecular interactions that specifically involve gliadins and glutenins. These proteins interact through both covalent and non-covalent bonds, which are essential for developing the viscoelastic properties of dough. Among the covalent interactions, disulfide bonds (S–S bridges) play a critical role [11]. These bonds are formed through the oxidation of cysteine residues present in the protein structure. In gliadins, which are monomeric proteins, disulfide bonds are predominantly intrachain, contributing to their extensibility. In contrast, glutenins, particularly the HMW subunits, form both intrachain and interchain disulfide bonds. The interchain disulfide bridges in glutenins are especially important, as they facilitate the formation of large, high-molecular-weight protein polymers that contribute significantly to the elasticity, strength, and overall viscoelasticity of the gluten network [12]. In particular, the molecular weight distribution of glutenins is a critical factor in determining dough quality, as it reflects the extent of disulfide bonding within the gluten network. This distribution is influenced by genetic factors (e.g., presence of specific HMW subunits like Dx5, the HMW-GS/LMW-GS ratio, and the abundance of chain terminators), environmental conditions (such as sulfur deficiency, heat, and water stress), and the redox environment during dough mixing, where reducing or oxidizing agents regulate disulfide bond formation and rearrangement. Together, these factors modulate gluten polymer size and structure, ultimately shaping dough functionality and rheological properties [13,14].
Additionally, non-covalent interactions such as hydrogen bonding—primarily due to the high glutamine content in gluten proteins—as well as intramolecular hydrogen bonds, hydrophobic interactions, Van der Waals forces, and ionic bonds, further stabilize the gluten structure and modulate its functional properties during mixing and baking [12].

2.2. Gluten-Related Proteins in Non-Wheat Cereals

The corresponding forms of gluten proteins in wheat (8–12% of flour) are also present in barley (~4%), rye (~3%), and oats (<2%), where they are called hordein, secalin, and avenin, respectively [15]. Due to the presence of celiac-active polypeptides, these proteins can trigger an immune-mediated inflammatory response in patients with CD [16,17].
Specifically, ω-secalins, HMW-secalins, γ-75k-secalins, and γ-40k-secalins in rye and β-hordeins, C-hordeins, D-hordeins, and γ-hordeins in barley, can be classified into three different groups according to their homologous (gliadins) amino acid sequences and MWs in the LMW group, medium-molecular-weight group, and HMW group. The amino acid sequences of these proteins feature repetitive motifs—such as QQPGQG, YYPTSP, or short repeats like QQP and QPG. Variations between these proteins and wheat gliadins often involve single amino acid substitutions or rearrangements of these repetitive units along the polypeptide chain, resulting in differences in their MWs. For example, the repetitive motifs for ω1,2-gliadins (QPQQPFP) are characteristic both of ω-secalins and C-hordeins. The MWs of LMW proteins—which include monomeric proteins such as α-gliadins, γ-gliadins, γ-40k-secalins, and γ-hordeins, as well as polymeric proteins like LMW glutenin subunits (LMW-GS), γ-75k-secalins, and β-hordeins—generally range from 28 to 35 kDa. An exception is γ-75k-secalins, which exhibit a higher MW of approximately 50 kDa. These proteins are characterized by unique repetitive motifs, such as QQPQQPFP found in γ-75k-secalins and β-hordeins, and are also present in wheat γ-gliadins. These repetitive sequences are key contributors to CD immunoreactivity in wheat and, similarly, are implicated in the pathogenic potential of rye and barley, as most CD-active peptides originate from these conserved motifs [18,19,20]. In contrast, avenins in oats are present in much lower amounts (<2% of flour) and generally exhibit fewer and shorter repetitive motifs. While some avenin peptides can elicit immune responses in a subset of CD patients, their overall immunogenic potential is lower compared to wheat, rye, and barley prolamins, consistent with the lower prevalence of avenin-related celiac reactivity [2].

3. Gluten Toxicity: Health Disorders Associated with Gluten Ingestion

3.1. Pathogenesis of CD

CD is a complex autoimmune disorder triggered by the ingestion of gluten in genetically predisposed individuals who express specific human leukocyte antigen (HLA) class II haplotypes, namely HLA-DQ2 and HLA-DQ8. Two main conditions are necessary for the development of CD: the presence of a genetic predisposition and the consumption of gluten [21]. The primary environmental risk factor for CD is the ingestion of gluten and related prolamins, which possess strong immunodominant properties in genetically susceptible individuals. Upon ingestion, gluten proteins are only partially hydrolyzed by gastrointestinal proteases, resulting in the formation of peptides that are highly resistant to further enzymatic degradation. This resistance is primarily due to the unique amino acid composition of gluten proteins—especially their high content of proline and glutamine residues arranged in repetitive motifs. These structural features render gluten peptides resistant to cleavage by key human gastrointestinal enzymes, including pepsin, trypsin, chymotrypsin, carboxypeptidases A and B, elastases, and brush-border membrane enzymes of the small intestine [22]. Notably, these enzymes exhibit limited activity at peptide bonds adjacent to proline residues, allowing the persistence of relatively large peptides (often nine or more amino acids in length). These peptides can cross the intestinal epithelium via paracellular or transcellular pathways and accumulate in the lamina propria. Gluten-derived peptides, rich in glutamine, are a good substrate for tissue transglutaminase (TGase), an enzyme released during inflammation. TGase deamidates these peptides, increasing their affinity for HLA-DQ2 and HLA-DQ8 molecules expressed on antigen-presenting cells in the gut. Once bound to these HLA molecules, the deamidated gliadin peptides are presented to CD4+ T cells, triggering a gliadin-specific immune response. This immune activation leads to intestinal inflammation and subsequent tissue damage characteristic of CD [22,23].
Extensive studies have identified several disease-driven gluten T cells epitopes, mainly derived from wheat—particularly from α-, γ-, and ω-gliadins as well as from glutenins—and, to a lesser extent, from barley and rye through their specific prolamins, hordeins, and secalins [24,25,26]. Nevertheless, many gluten T cell epitopes remain unidentified. Immunogenic sequences were identified using different approaches, ranging from mass spectrometry analysis of peptide fragments [27,28,29] to tagging of glutamine residues by TGase [30,31] to homology searches in edible cereal proteins [32], or by detection of gluten-specific T cells in the peripheral blood of CD patients after short-term consumption of gluten-containing food [32,33]. All the peptides identified contain one or more epitope sequences recognized by HLA-DQ2 and HLA-DQ8 molecules [33] after TGase-mediated deamidation [8,10,22]. A detailed list of CD epitopes and their nomenclature has been compiled by Sollid et al. [25], while a comprehensive repertoire of gluten peptides recognized by celiac T cells has been reviewed by Camarca et al. [24].
Several studies have evaluated peptides resistant to gastrointestinal hydrolysis using in vitro digestion of gliadins or gluten-containing foods. These investigations led to the identification of immunogenic gastro-resistant peptides (GIP), which include one or more epitope sequences [34,35,36,37]. Among these, the most emblematic is the 33-mer peptide (LQLQPFPQPQLPYPQPQLPYPQPQLPYPQPQPF), located in the N-terminal region 57–89 of α-gliadins. This fragment contains six copies of three different T-cell epitopes (DQ2-α-I, DQ2-α-II, and DQ2-α-III) and is characterized by a remarkable resistance to gastrointestinal proteolytic digestion, which amplifies its immunogenic potential [22]. In γ-gliadins, by contrast, the distribution of immunogenic sequences is more heterogeneous, with numerous stimulatory epitopes spread throughout the protein sequence. Among them, the 26-mer peptide (FLQPQQPFPQQPQQPYPQQPQQPFPQ) is particularly relevant, along with several shorter fragments containing immunogenic motifs such as QQPFPQQPQ [37]. A distinct category of gliadin peptides comprises the so-called ‘innate peptides’, which are not recognized by CD4+ T cells but are able to elicit an innate immune response in antigen-presenting cells (APC) such as monocytes, macrophages, and dendritic cells. The most studied peptide in this category is the α-gliadin p31–43 (or p31–49) peptide [38]. This peptide, or fragments including this sequence, has been shown to be highly resistant to in vitro gastrointestinal hydrolysis [39,40]. More recently, in vivo studies have revealed its detection in urine, confirming its remarkable resistance to proteolysis, intestinal permeability, and systemic circulation up to renal excretion [41].
In genetically predisposed individuals, the consumption of gluten or gluten-related proteins can trigger an aberrant immune response that leads to the development of symptoms associated with CD [2]. However, additional susceptibility factors beyond genetics have also been implicated. Alterations in the intestinal microbiota—specifically, reduced microbial diversity and shifts in microbial composition—have been associated with an increased risk of CD. Furthermore, emerging evidence suggests that certain viral infections may act as environmental triggers, contributing to the onset of the disease [42]. Clinically, CD is characterized by a range of symptoms primarily associated with malabsorption, including chronic diarrhea, steatorrhea, fatigue, weight loss, anemia, dermatological manifestations, and, in children, growth failure [2]. There is no defined minimum threshold of gluten that causes adverse effects, as this depends on individual patients. However, the scientific community recognizes that most CD subjects can tolerate trace amounts below 20 ppm, which is also the regulatory threshold for labeling a food as gluten-free [43]. To date, the only effective treatment for CD is a lifelong adherence to a strict gluten-free diet, which typically leads to symptom resolution and mucosal healing [44].

3.2. Non-Celiac Gluten Sensitivity

Non-celiac gluten sensitivity is a wheat-related condition that is neither IgE-mediated and not associated with an autoimmune response [45]. Determining the prevalence of NCGS is challenging due to the absence of validated biomarkers. Moreover, since the offending components may include other wheat constituents beyond gluten—such as amylase–trypsin inhibitors and fermentable carbohydrates (FODMAPs)—many experts now favor the broader term “non-celiac wheat/gluten sensitivity” [21]. Amylase–trypsin inhibitors (ATIs) are highly digestion-resistant proteins that can activate innate immunity and amplify symptoms [46]. FODMAPs may also provoke gastrointestinal distress; in some patients, they seem to be the main trigger, as low-FODMAP diets often bring marked relief [21]. It has been demonstrated that ancient diploid wheat (Triticum monococcum) activates immune cells far less than modern tetraploid or hexaploid varieties, suggesting it may be better tolerated by NCGS sufferers. Likewise, Tritordeum—a modern hybrid of durum wheat and barley—contains relatively little immunogenic gliadin and has been proposed as another potentially safe alternative [47,48].
Clinical symptoms of NCGS are generally less severe than those observed in CD or IgE-mediated wheat allergy [21]. Common manifestations include abdominal pain, bloating, gas, and diarrhea. Although these symptoms can resemble those of CD or wheat allergy, individuals with NCGS do not exhibit IgA anti-tissue transglutaminase autoantibodies or specific IgE antibodies against wheat proteins. Unlike CD, NCGS does not usually cause permanent intestinal damage, and symptoms generally improve or resolve with a gluten-free diet or reduced gluten intake [49]. While a minimum safe gluten threshold has been established for CD patients, no comparable value can be defined for individuals affected by NCGS, as their reactions are highly variable and dose-dependent [50].

3.3. Wheat Allergy

Wheat allergy is an adverse immune response triggered by either IgE-mediated or non-IgE-mediated mechanisms. Two distinct types of IgE-mediated allergic diseases related to wheat and other cereals have been identified. The first type includes occupational respiratory allergies, such as baker’s asthma and rhinitis, which result from inhaling flour particles in environments like bakeries. The second type refers to a true IgE-mediated food allergy to cereals. This process begins with the sensitization phase, where the immune system produces IgE antibodies specific to wheat allergens. Upon subsequent exposure, these allergens (e.g., wheat proteins) bind to IgE antibodies on the surface of mast cells and basophils, causing them to activate. This activation leads to the release of pro-inflammatory mediators such as histamine, which are responsible for the clinical symptoms of allergy [51,52,53].
According to the Allergen Nomenclature Sub-Committee of the International Union of Immunological Societies (IUIS), 28 distinct wheat allergens have been identified to date [54]. These allergens are present in both the water-insoluble fraction—comprising gliadins and glutenins—and the salt-soluble fraction, which includes lipid transfer proteins (LTPs) and α-amylase inhibitors (ATIs). Both ATIs and LTPs, such as Tri a 14, are heat-stable and are frequently recognized by IgE antibodies in patients with severe reactions to wheat, including wheat-dependent exercise-induced anaphylaxis (WDEIA) and baker’s asthma. Another heat- and digestion-resistant allergen, Tri a 37, a plant defense protein, has also been associated with severe allergic responses to wheat. Additionally, ω5-gliadin, designated as Tri a 19, has been strongly implicated in WDEIA and anaphylactic reactions to wheat, particularly in children [55,56].
The clinical symptoms of wheat allergy vary widely. They may present as typical food allergy symptoms affecting the skin, gastrointestinal tract, or respiratory system. In rare cases, a severe condition known as WDEIA may occur. This form is marked by respiratory distress, urticaria, throat tightness, and a rapid drop in blood pressure, typically following wheat ingestion combined with physical activity [51,56]. In IgE-mediated wheat allergy, allergic reactions are dose-dependent and vary between individuals. The exact threshold depends on the patient’s sensitivity [50].
Currently, the primary approach to managing wheat allergy involves strict avoidance of wheat-containing products. There is, however, no validated strategy for primary prevention. Developing hypoallergenic wheat-based foods with reduced allergenic proteins may offer a promising preventive option and help lower the incidence of wheat allergies in the future.

3.4. Detection and Quantification of Gluten Peptides

For individuals with gluten-related disorders, gluten must be strictly avoided in the diet, including even trace amounts that may result from cross-contamination, which is particularly critical for CD patients. Cross-contamination can occur at various stages of the food supply chain, including harvesting, transportation, storage, and processing, when gluten-free grains come into contact with gluten-containing cereals. Additionally, shared equipment or facilities in food manufacturing further increase the risk of unintentional gluten contamination.
Given these challenges, the development and application of reliable methods for the detection and quantification of gluten peptides is of paramount importance. Different investigative techniques are employed to determine gluten peptides, including immunochemical-based methods (ELISA, lateral flow devices) and proteomics-based methods (mass spectrometry) [57]. These methods differ significantly in terms of cost, sensitivity, and specificity. Nevertheless, they must be sensitive enough to detect trace levels of gluten, specific enough to avoid false positives, and robust enough to be applied across a wide variety of food matrices [58].
ELISA is a widely used immunochemical method for the detection and quantification of gluten in raw and processed foods, as well as beverages. Its ease of use and rapid results make it the most common method for gluten analysis in the food industry. Commercial ELISA kits typically employ monoclonal antibodies such as R5 and G12 [57]. The Codex Alimentarius standard for gluten detection officially endorses the use of the R5 monoclonal antibody, which recognizes specific peptide epitopes (QQPFP, QQQFP, LQPFP, and QLPFP) present in α-/β-, ω-, and γ-gliadins, not only in wheat but also in rye (secalin) [59]. The G12 monoclonal antibody was developed to target the immunotoxic 33-mer peptide of α2-gliadin. G12 specifically binds to the QPQLPY epitope, one of the most immunogenic fragments of the 33-mer [60]. Novel monoclonal antibodies that specifically recognize deamidated gliadin have also been proposed for the detection of gluten proteins that may be modified during food processing [60,61].
Sandwich ELISA is the commonly used format for detecting gluten contamination from intact proteins, as it requires two distinct epitopes on the target molecule for antibody binding. However, it is not effective for detecting hydrolyzed gluten, where proteins are broken down and epitopes may be lost. In contrast, competitive ELISA is more suitable for detecting small antigens that possess only a single epitope, such as hydrolyzed gluten fragments. Competitive ELISAs based on R5 and G12 antibodies are commonly used for this purpose [62,63].
Immunochemical assays have also been developed not only for the detection of gluten in food but also for the analysis of gluten immunogenic peptides in biological fluids, in order to monitor dietary intake and gluten exposure. An ELISA based on G12 has been developed for the detection of trace gluten in feces [64]. Lateral flow devices are rapid, strip-based tests that detect prolamin epitopes to estimate gluten content, similar to ELISA methods, generally based on G12 and A1 antibodies [65,66]. They are commonly used for onsite testing within manufacturing facilities as part of control programs and to support hazard analysis and critical control point (HACCP) systems—from raw material storage to final products. LFDs offer key advantages, including user-friendliness, rapid results, and the convenience of on-site analysis [64,65,66,67].
Mass spectrometry-based proteomic analysis has been applied for the identification and quantification of immunoreactive cereal proteins [55] and is increasingly used as an alternative or complementary approach to immunochemical methods. This approach provides not only qualitative and quantitative results but also a detailed characterization of the amino acid sequences of gluten peptides [68,69,70].
These methods are based on the release of immunogenic peptides following enzymatic treatment, followed by their detection and quantification. Typical targets include α- and γ-gliadin-derived peptides, and often the proteolytically resistant 33-mer peptide [68,69,70], which is believed to play a pivotal role in the pathogenesis of CD [22,37,68,71]. Thanks to its versatility, this technique has been proposed for the analysis of gluten in foods as well as for the detection of gluten immunogenic peptides in biological samples. In particular, a recent study identified gluten peptides in subjects after the ingestion of gluten-containing foods. Urine peptidomic analyses revealed high variability in gluten peptides between subjects, which may be due to interindividual differences in protein digestion and gastrointestinal damage [41,72,73].

4. Beneficial Aspects of Gluten-(Derived Peptides)

Beyond their well-known role in the structure/functionality of gluten as well as their involvement in CD, wheat proteins are also a source of bioactive peptides with potential beneficial effects on human health [74,75]. These peptides are inactive within the native protein sequence but can be released through enzymatic hydrolysis, either during gastrointestinal digestion, food processing such as fermentation or germination, or through the action of microbial proteases [76,77]. Several bioactive peptides have been identified from wheat proteins, particularly from gliadins and glutenins, but also from non-gluten proteins like albumins and globulins, and they exhibit a wide range of physiological functions [78,79].
Among the most studied activities is the antioxidant potential of certain peptides capable of scavenging free radicals and inhibiting lipid peroxidation, thereby contributing to the reduction of oxidative stress—a key factor in the development of chronic inflammatory diseases. These antioxidant peptides are typically released from albumins and globulins following germination or fermentation and include sequences such as AREGETVVPG, short peptides like GPF, GPE, and FGE (with molecular weights < 1.5 kDa), as well as others like YDWPGGRN, TGP, and QPYPQQPQ, which may also exert protective effects against oxidative stress in intestinal cells such as Caco-2 and possibly display anti-aging properties [39]. In addition, several wheat oligopeptides such as LY, PY, YQ, APSY, RGGY, and LVS have demonstrated both antioxidant and antihypertensive activities, supporting their potential use in nutraceutical applications [80].
Another notable functional category includes peptides with angiotensin-converting enzyme (ACE) inhibitory activity, which may help reduce blood pressure by modulating the renin-angiotensin system. For instance, the tripeptide IAP, derived from gliadin hydrolysates, exhibits potent ACE-inhibition effects and has shown efficacy in lowering blood pressure in hypertensive rats. Other wheat-derived peptides, such as VPL, WL, and WP, also demonstrate ACE-inhibitory effects, largely due to their hydrophobic amino acid content, especially proline and tryptophan. Asoodeh et al. [81] identified sequences like IPALLKR and AQQLAAQLPRMCR from wheat gluten hydrolysates, showing that ACE-inhibitory activity is strongly influenced by peptide structure, particularly the presence of hydrophobic and aromatic residues such as tryptophan, tyrosine, phenylalanine, and proline in the C-terminal region. Similarly, studies conducted on isolated peptides from wheat gluten for their potential use as ACE inhibitors emphasize the importance of generating gluten hydrolysates to enhance their health-promoting properties—an approach that may be particularly relevant for celiac individuals, provided that the hydrolysates are rendered non-immunogenic [82].
In vivo studies have demonstrated that wheat peptides, administered as dietary supplements, exert significant hypotensive and antioxidant effects. Experiments conducted on hypertensive rat models showed that hydrolyzed wheat gluten proteins and certain natural peptides derived from wheat are able to safely reduce blood pressure by inhibiting angiotensin production. In addition, wheat peptides exhibited marked antioxidant properties [82].
Similarly, in vivo experiments on animal models demonstrated that a mixture of corn and wheat peptides can prevent the onset of diabetes in NOD (non-obese diabetic) mice [83]. Moreover, wheat peptides have also been shown to protect against gastric damage induced by NSAIDs (Non-Steroidal Anti-Inflammatory Drugs) in rats, by reducing the oxidative stress associated with these drugs [84].
Another fascinating class of wheat-derived peptides is represented by gluten exorphins—opioid-like peptides released from gliadins during enzymatic digestion—which are capable of binding to opioid receptors and may influence mood, appetite, and gastrointestinal motility. While their pharmacological activity has been well documented in vitro, their physiological relevance in vivo remains debated. Key examples include gluten exorphin A4 (QYYP), A5 (GYYPT), B4 (YGGW), B5 (YGGWL), C5 (YPISL), and gliadorphin-7 (YPQPQPF) [81,82]. To exert their activity, these peptides would need to cross the intestinal barrier, reach the systemic circulation, and remain intact long enough to access the central nervous system—a condition that has not yet been clearly demonstrated. In such cases, gluten exorphins may exert effects on the central nervous system, potentially influencing behavior, attention, and emotional regulation [85,86,87,88]. Some hypotheses suggest a possible link between gluten-derived exorphins and neurological or neurodevelopmental disorders, such as autism spectrum disorders or schizophrenia, although conclusive clinical evidence is still lacking [85,86,87,88,89,90]. Therefore, while gluten exorphins may have interesting pharmacological properties, their potential role in adverse physiological or neurological effects cannot be excluded and warrants further investigation.
In addition to their antioxidant, antihypertensive, and opioid-like activities, certain wheat-derived peptides have demonstrated immunomodulatory properties by influencing cytokine production [91]. Furthermore, there is emerging evidence supporting the hypocholesterolemic and antidiabetic potential of specific peptides that inhibit key enzymes involved in carbohydrate and lipid metabolism, such as α-glucosidase and pancreatic lipase, suggesting their potential usefulness in managing metabolic syndrome [92,93].
It is important to note that the bioactivity and physiological relevance of these peptides depend largely on their bioavailability, particularly their resistance to gastrointestinal digestion and their ability to reach target tissues intact [94,95]. In this context, hydrolysis with selected enzymes has proven to be a promising strategy to enhance the release, stability, and functionality of bioactive peptides, especially in the development of functional wheat-based foods designed to support health beyond basic nutrition (Figure 1).

5. Strategies to Minimize Gluten Toxicity While Enhancing Health Benefits

While adherence to a strict, lifelong gluten-free diet remains the only effective treatment, alternative strategies, in order to mitigate adverse immune responses in susceptible individuals—such as reducing the gluten content in food and beverages—are also being explored [96,97,98,99].
Over the last decade, several studies have focused on reducing or eliminating gluten toxicity by using methodologies based on biological measurements, genetic approaches, or a combination of biotechnological and non-biotechnological techniques (Figure 2). The objective was to apply non-invasive enzymatic strategies in order to hydrolyze immunogenic gluten peptides, while simultaneously enhancing, where feasible, the nutritional profile and organoleptic characteristics of the food matrix [100].
The use of proteolytic enzymes capable of degrading immunogenic peptide sequences rich in proline and glutamine is one of the paths to follow, thereby preventing the ingestion of peptides that elicit autoimmune responses in individuals with celiac disease. These enzymes, in fact, can be either exogenous—typically of microbial origin as lactic acid bacteria (LAB)—or endogenous, involved in natural metabolic processes during grain germination and sprouting. Endogenous activity may also include enzymes produced by microorganisms associated with the plant microbiome, such as fungal xylanases, which contribute to the breakdown of complex macromolecules during the early developmental stage. Table 1 provides a detailed summary of the degradation strategies implemented, the specific enzymes involved, and the conditions under which each method was applied.

5.1. Fermentation with Bacteria Strains

Fermentation with LAB is one of the most widely used biotechnological strategies to reduce the toxicity of gluten through microbial enzymatic activity. Numerous studies [143,144] have demonstrated that sourdough fermentation of wheat or gluten-containing flours reduces IgE recognition and that specific LAB starter cultures can partially degrade gluten proteins resistant to digestion. This enzymatic degradation may enhance gastrointestinal digestion and reduce sensitization risk by limiting exposure to intact immunogenic epitopes. However, achieving a truly gluten-free product using LAB cultures remains a significant challenge.
Pioneering work by Di Cagno et al. [131,132] identified the proteolytic capabilities of selected LAB strains—including Lactobacillus alimentarius 15M, Lactobacillus brevis 14G, Lactobacillus sanfranciscensis 7A, Lactobacillus plantarum CF1, and Lactobacillus hilgardii 51B—toward gliadin-derived peptides implicated in CD. These strains significantly degraded proline-rich sequences resistant to gastrointestinal proteases. Additionally, their use enhanced the nutritional quality of gluten-free bread formulations, demonstrating both detoxifying and nutritional benefits [101,133,145,146].
The application of probiotic LAB strains—such as Streptococcus thermophilus, Lactobacillus plantarum, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus delbrueckii subsp. bulgaricus, Bifidobacterium breve, Bifidobacterium longum, and Bifidobacterium infantis—have also been explored to reduce immunogenic gluten peptides in wheat dough. While these strains exhibit proteolytic activity, particularly during extended fermentation, they remain among the least effective strategies for achieving substantial gluten degradation [134,135,137].
Although sourdough-based fermentation does not lead to complete degradation of toxic gluten fragments, it contributes significantly to the improvement of texture, palatability, aroma development, and reduction of antinutritional factors in baked products [137].
Recently, Lactococcus lactis LLGKC18 was evaluated using a functional rat basophil leukemia (RBL) cell model activated with sera from allergic patients. The study showed significantly reduced RBL degranulation, indicating decreased allergenicity, despite the presence of residual peptides. MS/MS analysis confirmed the degradation of several immunogenic peptides, suggesting that Lactococcus lactis LLGKC18 produces proteases capable of targeting immunodominant sequences of ω5-gliadin [136].
Furthermore, various Bacillus species—including Bacillus subtilis, Bacillus tequilensis, and Bacillus cereus QAUSD07—have been shown to produce peptidases and proteases, such as subtilisin and thermolysin, that hydrolyze gliadin into smaller peptides and amino acids. Notably, proteases from Bacillus subtilis exhibit high specificity toward proline-rich regions, effectively degrading immunogenic peptides such as the 33-mer [102,147].
Recent advances in food processing technologies have increasingly targeted the elimination of toxic gluten epitopes through combined biotechnological strategies. One notable approach involves the use of fungal proteases in combination with probiotic Lactobacillus strains, particularly Lactobacillus sanfranciscensis, during extended sourdough fermentation [148]. This method has proven effective in vitro, achieving gluten degradation to levels below 10 ppm, which meets the threshold for gluten-free labeling and significantly reduces immunogenic potential [102,147,148].
A synergistic formulation of probiotic strains—including Lactobacillus casei LC130, Lactobacillus paracasei LPC100, and Streptococcus thermophilus ST250—has demonstrated the ability to hydrolyze immunoreactive gliadin peptides. This enzymatic activity holds promise for mitigating the effects of inadvertent gluten exposure, such as contamination in gluten-free foods, offering potential benefits for individuals with gluten-related disorders [134,135,138,148].
Additional evidence suggests that sourdough fermentation, combining yeast and LAB, further reduces gluten immunogenicity by promoting gluten depolymerization and hydrolysis, particularly through weakening glutenin polymerization and breaking down glutenin-derived peptides. Fermentations involving Lactobacillus brevis and Pediococcus pentosaceus, in combination with yeast, have shown a significant reduction of toxic gluten proteins in wholemeal products [134,135,138,148].
Importantly, the application of LAB in vivo-like settings has also been explored. A recent study by Nikoloudak et al. [149] evaluated a novel formulation combining probiotic strains with commercial proteolytic enzymes under simulated gastrointestinal conditions. The treatment successfully degraded gluten to below 20 ppm, the accepted safety threshold, indicating its strong potential for mitigating gluten-related immunotoxicity in clinical contexts.

5.2. Combined Enzymatic Strategies: Germination Meets Microbial and Fungal Enzyme Applications

The application of glutanase, particularly those derived from plants, fungi, and other microorganisms, and often associated with endogenous protease, has seen significant advancement due to their capacity to function both in vivo and ex vivo in effectively hydrolyzing gluten. This enzymatic activity facilitates the degradation of immunogenic epitopes that are degraded or completely destroyed, thereby reducing the toxicity associated with gluten peptides [73,129].
One of the principal mechanisms to obtain gluten hydrolysis is the activation of endogenous proteases in grain by germination. In barley, for example, this physiological process induces the activation of specific cysteine proteases, which are capable of hydrolyzing immunotoxic amino acid sequences within hordein, the predominant prolamin storage protein [129]. Recent studies have also examined wheat gluten degradation during germination, revealing that endoprotease activity begins around day 3 and reaches its peak by day 7. These findings suggest that germination under controlled conditions can effectively reduce gluten immunogenicity. However, extended germination increases amylase activity, which may lead to technological issues [103,104,130].
In addition to germination, other approaches have explored enzymatic modification of gluten using peptidases as transferases. One study demonstrated that binding methionine to gluten using α-chymotrypsin could alter its structure, offering a novel strategy for reducing its immunoreactivity [103].
Other approaches to obtain low content gluten products are to use other types of barley cysteine proteases, as the papain-like C1A family and the C13 family, also called legumains, that degrade completely epitopes eliciting CD [105,106]. These classes of endoproteases are also present in plants. Notably, papain, bromelain, and actinidin—classified as cysteine endoproteases—exhibit significant hydrolytic activity. Papain has been utilized in wheat gluten hydrolysates, a by-product of wheat starch production, effectively disrupting the gluten protein structure. Bromelain has been employed to produce hypoallergenic wheat flour, as it facilitates the cleavage of peptide bonds adjacent to proline residues, which are typically resistant to proteolytic degradation. Actinidin, naturally found in kiwifruit, papaya, and pineapple, enhances gluten digestibility by promoting proteolysis during gastrointestinal digestion, thereby improving protein bioavailability in the small intestine [107].
More generally, promising enzymatic approaches using plant proteases have been explored to reduce gluten allergenicity through targeted hydrolysis. Protease extracts purified from Nigella sativa effectively hydrolyze gluten and gliadin, suggesting their potential for detoxifying gliadin proteins. Similarly, neprosin, a protease derived from Nepenthes pitcher plants, has shown selective activity toward gliadin. Additionally, Cuminum cyminum (cumin) latex has been reported to enhance pepsin’s proteolytic activity; however, its application in gluten hydrolysis has not yet been evaluated. Another promising candidate is caricain, extracted from Carica papaya latex, which is able to detoxify gliadin in whole wheat flour, supporting its potential as a natural protease for gluten degradation [105,106,107]. Furthermore, the combination of a microbial protease (alcalase) with a plant protease (papain) demonstrated significantly greater effectiveness in degrading gliadin compared to the use of either enzyme alone in wheat flour. As a result, the resulting flour extract exhibited substantially reduced allergenic properties, outperforming hydrolysates produced using individual enzymes such as chymotrypsin, flavourzyme, trypsin, or pepsin [107].
Recent scientific studies increasingly employ a combination of endoproteases and exoproteases from different matrices to enhance the efficiency and specificity of gluten protein degradation [101]. The combination of a prolyl endopeptidase from Flavobacterium meningosepticum and a glutamine-specific endoproteinase from barley showed a detoxifying capacity of immunogenic gluten peptides during simulated gastrointestinal digestion. Another method is the synergistic employment of the prolyl endoproteases from Aspergillus niger (AN-PEP) and from Sphingomonas capsulata (SC-PEP), in conjunction with EP-B2, a cysteine endoprotease derived from barley, which has demonstrated enhanced efficacy in degrading proline-rich peptides, facilitating the breakdown of protease-resistant gluten peptides associated with CD pathogenesis. Furthermore, the cysteine endoprotease EP-B2 has also been extended to the brewing industry, employed to enzymatically detoxify hordeins, permitting the production of gluten-free beer from conventional cereals and pseudocereals while maintaining sensory and functional characteristics [108].
An innovative enzymatic approach for reducing the immunogenicity of gluten in CD involves the application of microbial transglutaminase (mTG), an endo-γ-glutamine:ε-lysine transferase. This enzyme catalyzes transamidation reactions, forming isopeptide bonds between the γ-carboxamide group of glutamine residues in gliadin and primary amines such as lysine or n-butylamine. Importantly, this targeted modification of gliadin peptides has been shown to prevent the formation of immunodominant T-cell epitopes, thereby inhibiting interferon-γ (IFN-γ) production by intestinal T cells from CD patients. These findings highlight transamidation as a promising technological intervention for developing wheat-based products with reduced immunogenic potential, offering a potential pathway toward safer dietary options for individuals with CD [109,110].

5.3. The Oral Therapy

Enzyme supplement therapy using either bacterial or fungal endopeptidases or endoproteases from germinating cereals has been suggested to promote the hydrolysis of gluten proteins and destroy toxic peptides. A prerequisite is that such enzymes are first safe for human consumption and then obviously active under gastro-duodenal conditions. Several enzymes, including prolyl endopeptidases, cysteine proteases, and subtilisins, can hydrolyze the human digestion-resistant gluten peptides both in vitro and in vivo [37].
EP-B2, a glutamine-specific endoprotease active during barley seed germination and commonly used in enzymatic treatments, remains active under gastric conditions (low pH), is resistant to pepsin, but is hydrolyzed at physiological concentrations of trypsin. It exhibits strong specificity for the QXP sequence, which is abundant in the immunotoxic 33-mer and other gluten-derived peptides. These properties make EP-B2 a promising candidate for use as a gastric-active enzyme in CD [111].
Proline endoproteases (PEPs) have been investigated as potential oral glutenases for CD therapy [112,113]. These serine proteases cleave peptide bonds at the carboxy end of proline residues, a common feature of immunogenic gluten peptides. Microbial sources such as Flavobacterium meningosepticum (FM-PEP), Sphingomonas capsulata (SC-PEP), and Myxococcus xanthus (MX-PEP) have been studied for this purpose [37]. Among them, SC-PEP shows the greatest promise due to its stability across acidic pH levels and resistance to pepsin degradation, unlike FM-PEP and MX-PEP, which are susceptible to digestion by pepsin and bile salts.
Another promising enzyme is a prolyl endopeptidase from the fungus Aspergillus niger, named AN-PEP, which is industrially scalable, active at gastric pH (3–5), and resistant to pepsin, similar to SC-PEP. Although less specific for immunodominant gluten epitopes, AN-PEP efficiently degrades gliadin into smaller peptides and may enhance gluten detoxification when used in combination with more selective enzymes, such as dipeptidyl peptidase-IV (DPP-IV) [23]. DPP-IV occurs naturally in small amounts in the small intestinal brush border; however, it has been obtained commercially from the fungus Aspergillus oryzae. It acts on the amino-terminal side to liberate X-Pro dipeptides of gliadin, and it works best at neutral pH; hence, it starts action only in the intestine, and it exploits a limited proteolytic effect [75,101].
Another fungal enzyme from Aspergillus niger, the aspergillopepsin (ASP), has demonstrated high efficiency in degrading CD–active peptides. With an optimal activity at pH 3.0, ASP can extensively hydrolyze dietary gluten into short peptides. However, it lacks specificity for the immunodominant 33-mer peptide of α2-gliadin. ASP is considered safe for human consumption and may be used in combination with more specific and potent gluten-degrading enzymes, such as EP-B2 or microbial prolyl endopeptidases, to enhance overall therapeutic efficacy [108,114].
A cysteine protease from the papain family, Triticain-α from Triticum aestivum, has been demonstrated to efficiently cleave α-, γ-, and ω-gliadin and glutenin, exhibiting an optimal enzymatic activity under acidic conditions (gastric phase). However, under intestinal conditions and in the presence of trypsin, this enzyme becomes susceptible to inactivation. Therefore, its functional activity is likely limited to the gastric phase of gluten digestion, prior to intestinal transit [115].
Recently, E40, a bacterial protease derived from Actinoallomurus sp. A8 has shown high efficacy in degrading the immunodominant 33-mer peptide and whole gliadin proteins, with optimal activity across a pH range of 3–6. Recombinant production of E40 using Streptomycetes—recognized as a safe protein source for human consumption—further supports its suitability for dietary management of gluten-related disorders. E40 is resistant to degradation by both pepsin and trypsin and retains activity in the acidic environment of the stomach. In simulated gastrointestinal conditions, E40 effectively digests gluten without releasing immunotoxic peptides, thereby preventing the downstream activation of the inflammatory cascade characteristic of CD. Its efficacy has also been validated in various food matrices, including beer, bread, and pasta. Notably, gluten levels in E40-treated samples consistently fell at or below the 20 ppm threshold required for gluten-free labeling; in fact, only trace amounts of immunogenic peptides derived from ω- and γ-gliadins were observed. For these reasons, E40 has been proposed as a promising candidate for oral enzymatic therapy aimed at managing gluten toxicity [116,117].
Again, Kuma030, an engineered endopeptidase derived from the microbial protease kumamolisin of Alicyclobacillus sendaiensis, has demonstrated the ability to degrade a broad range of immunogenic gliadin epitopes under gastric conditions (optimal activity at pH 4). Its enzymatic properties and substrate specificity highlight its potential as a promising candidate for the enzymatic treatment of CD [118,119].
While these enzymes have demonstrated glutenase activity under optimal conditions, it remains unclear whether they fully eliminate all immunogenic epitopes and prevent immune activation. Most have been tested only in small gluten challenge studies in patients on a gluten-free diet. Further clinical trials are needed to assess their efficacy and safety in real-world settings.

5.4. Genetic and Breeding Approaches

Another way to produce gluten-free wheat-based foods is by developing wheat varieties with fewer harmful CD epitopes that trigger CD. Achieving this requires a thorough understanding of the complexity of the wheat genome and the structure of the gene families encoding gliadin proteins that harbor CD epitopes. At the genomic level, the presence of CD epitopes varies among gliadin genes, across the homoeologous wheat chromosomes (A, B, and D), and among different wheat varieties and species. The α-gliadins encoded by the D genome contribute the highest number of CD epitopes, while those from the B genome contribute the fewest. Similarly, for γ-gliadins, the greatest number of CD epitopes has been identified in genes located on the D genome. Although less sequence information is available for ω-gliadins, their role in CD immunogenicity has recently been recognized due to T-cell cross-reactivity with related epitopes in rye and barley.
Genetic variations naturally present in different wheat cultivars offer opportunities to develop varieties that are safe for individuals with CD. Particular attention has been given to Triticum monococcum (einkorn), an ancient diploid wheat species regarded as a promising candidate for a species with reduced content of CD epitopes and with more digestible wheat gliadin [120,140,141]. Unlike modern wheat, einkorn lacks the D genome that encodes the highly immunogenic 33-mer gliadin peptide. Multiple in vitro and ex vivo studies have shown that einkorn exhibits a significantly lower potential to provoke immune responses in individuals with CD [120]. Although einkorn proteins still contain some immunogenic and toxic peptides, subtle variations in their amino acid sequences may reduce their harmful effects, suggesting a potentially lower overall toxicity compared to modern wheat [142].
Recent advances in biotechnology, particularly RNA interference (RNAi), have enabled the development of low-gluten wheat varieties. RNAi works by introducing complementary RNA molecules that bind to the target mRNA, promoting its degradation and thereby preventing the translation of specific genes, reducing gliadin expression and developing wheat lines with fewer gluten genes and/or gluten genes with inactivated CD epitopes [121,139].
Recently, targeted gene editing using CRISPR/Cas9 has been applied to gliadins. These methods produce offspring with silenced, deleted, and/or edited gliadins, which, overall, may reduce the exposure of patients to CD epitopes. The application of CRISPR-Cas9 silencing technology was also used to silence CM3 and CM16 ATI genes in durum wheat, in order to obtain transgenic lines that showed an effective decrease in the target gene coding for allergenic proteins [122,123,124].
Synthetic hexaploid wheat breeding represents a promising strategy for developing Triticum species with reduced gluten immunogenicity. This approach involves hybridizing Triticum turgidum ssp. durum with Aegilops tauschii lines with low levels of CD-toxic gliadins. The resulting SHW lines could combine low gluten immunogenicity with valuable agronomic traits. These lines would serve as key resources in pre-breeding programs, facilitating the development of commercial wheat cultivars that are both agronomically competitive and less immunoreactive for individuals with CD or NCGS [125].
Deletion lines of wheat, such as those derived from “Chinese Spring,” have been used to remove specific chromosomal regions, targeting both CD-epitopes and functional traits like dough quality. For example, deletion of the 6DS arm, which contains the 6D α-gliadin locus, significantly reduced Glia-α1 and Glia-α3 immunogenicity but resulted in stiffer, less elastic dough. In contrast, removing ω-gliadins, γ-gliadins, and LMW-glutenins from chromosome 1DS lowered certain CD epitopes without compromising dough properties. However, these modifications involve genetic alterations that persist in the wheat lines, limiting their commercial viability in regions with strict regulations on genetically modified organisms (GMOs) [126].
Recently, chemical strategies for reducing gluten toxicity have been applied. The methane sulfonate, for example, induces G/C to A/T nucleotides in the DNA transitions, often causing missense mutations in α- and γ-gliadin genes, including within CD epitopes [127]. Gamma irradiation, in contrast, causes DNA strand breaks, leading to deletions, substitutions, or insertions. This can disrupt or delete gliadin genes and associated epitopes [128].
In the future, low gluten immunogenicity may become one of several traits prioritized in commercial wheat programs. In order to produce coeliac-safe food products will require significant effort and likely necessitate adaptations in industrial grain and food processing methods. Summarizing, two complementary approaches may contribute to this goal: plant breeding, to develop wheat varieties with a reduced immunogenic gluten fraction, and food-processing innovations that modify or remove immunogenic gluten components during grain and product processing.

6. Conclusions and Future Perspectives

The bioavailability and in vivo effects of gluten-derived bioactive peptides represent a promising but still underexplored field of research with important implications for human health. While numerous in vitro studies have demonstrated that these peptides possess bioactive properties, it remains crucial to deepen our understanding of their bioavailability and biodisponibility through well-designed animal models and human clinical trials. Such studies will help clarify the real health benefits of these bioactive peptides and support their potential use as functional food components or nutraceuticals.
In the context of CD, a strict gluten-free diet remains the only effective and widely accepted therapeutic approach. Despite this, innovative strategies aimed at reducing gluten toxicity are gaining traction, holding great promise in improving the quality of life for genetically predisposed individuals and may eventually offer alternatives or adjuncts to the gluten-free diet.
Similar therapeutic avenues are being explored for NCGS, although this condition’s pathophysiology remains less understood. The identification of the specific gluten-derived or other wheat-related molecules responsible for triggering symptoms in NCGS is an ongoing area of research, and future studies are needed to define precise diagnostic biomarkers and targeted therapies.
Ultimately, gluten should not be viewed as inherently harmful for the general population, but rather as a dietary component contraindicated only for individuals with specific genetic predispositions or sensitivities. For the majority, gluten-containing foods can be part of a balanced and nutritious diet, potentially offering beneficial bioactive peptides that support health. The challenge for future research will be to unravel the complex dual nature of gluten proteins—harnessing their positive effects while minimizing risks for susceptible individuals—thereby guiding personalized nutritional recommendations and innovative therapeutic strategies.

Author Contributions

The authors’ responsibilities were as follows: L.D.S.: investigation, data curation, writing, review, and editing. G.M.: conceptualization, investigation, writing, supervision, review, and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This review was financially supported by “On Foods—Research and innovation network on food and nutrition Sustainability, Safety and Security—Working ON Foods”-National Recovery and Resilience Plan (NRRP), Mission 4 Component 2 Investment 1.3—Call for proposals No. 341 of 15 March 2022 of Italian Ministry of University and Research funded by the European Union—NextGenerationEU; Award Number: Project code PE00000003, Concession Decree No. 1550 of 11 October 2022 adopted by the Italian Ministry of University and Research; Project PRR.AP022.022; Spoke 3 “Food safety of traditional and novel foods” and Spoke 4 “Food quality and nutrition”.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
APCAntigen presenting cells
ATIAmylase–trypsin inhibitors
CDCeliac disease
FODMAPFermentable oligosaccharides, disaccharides, monosaccharides, and polyols
GMOGenetically modified organism
IFN-γinterferon-γ
kDaKilo Dalton
LABLactic acid bacteria
LTPLipid transfer protein
MWMolecular weight
NCGSNon celiac gluten sensitivity
NSAIDsNon-Steroidal Anti-Inflammatory Drugs
NODNon-obese diabetic
pIIsoelectric point
RBLRat basophil leukemia
SDS-PAGESodium Dodecyl Sulphate—PolyAcrylamide Gel Electrophoresis
WDEIAWheat-dependent exercise-induced anaphylaxis

References

  1. Shewry, P.R.; Halford, N.G. Cereal seed storage proteins: Structures, properties and role in grain utilization. J. Exp. Bot. 2002, 53, 947–958. [Google Scholar] [CrossRef]
  2. Caio, G.; Volta, U.; Sapone, A.; Leffler, D.A.; De Giorgio, R.; Catassi, C.; Fasano, A. Celiac disease: A comprehensive current review. BMC Med. 2019, 17, 142. [Google Scholar] [CrossRef]
  3. Besser, H.A.; Khosla, C. Celiac disease: Mechanisms and emerging therapeutics. Trends Pharmacol. Sci. 2023, 44, 949–962. [Google Scholar] [CrossRef]
  4. Esfandi, R.; Walters, M.E.; Tsopmo, A. Antioxidant properties and potential mechanisms of hydrolyzed proteins and peptides from cereals. Heliyon 2019, 5, e01538. [Google Scholar] [CrossRef] [PubMed]
  5. Mamone, G.; Di Stasio, L.; Vitale, S.; Picascia, S.; Gianfrani, C. Analytical and functional approaches to assess the immunogenicity of gluten proteins. Front. Nutr. 2023, 9, 1049623. [Google Scholar] [CrossRef] [PubMed]
  6. Shewry, P. Wheat grain proteins: Past, present, and future. Cereal Chem. 2023, 100, 9–22. [Google Scholar] [CrossRef]
  7. Shewry, P.R.; Belton, P.S. What do we really understand about wheat gluten structure and functionality? J. Cereal Sci. 2024, 117, 103895. [Google Scholar] [CrossRef]
  8. Shewry, P. What is gluten—Why is it special? Front. Nutr. 2019, 6, 101. [Google Scholar] [CrossRef]
  9. De Santis, M.A.; Giuliani, M.M.; Giuzio, L.; De Vita, P.; Lovegrove, A.; Shewry, P.R.; Flagella, Z. Differences in gluten protein composition between old and modern durum wheat genotypes in relation to 20th century breeding in Italy. Eur. J. Agron. 2017, 87, 19–29. [Google Scholar] [CrossRef]
  10. Wieser, H.; Koehler, P.; Scherf, K.A. Chemistry of wheat gluten proteins: Qualitative composition. Cereal Chem. 2023, 100, 23–35. [Google Scholar] [CrossRef]
  11. Kłosok, K.; Welc, R.; Fornal, E.; Nawrocka, A. Effects of Physical and Chemical Factors on the Structure of Gluten, Gliadins and Glutenins as Studied with Spectroscopic Methods. Molecules 2021, 26, 508. [Google Scholar] [CrossRef]
  12. Ooms, N.; Delcour, J.A. How to impact gluten protein network formation during wheat flour dough making. Curr. Opin. Food Sci. 2019, 25, 88–97. [Google Scholar] [CrossRef]
  13. Wieser, H. Chemistry of gluten proteins. Food Microbiol. 2007, 24, 115–119. [Google Scholar] [CrossRef]
  14. Bonomi, F.; Iametti, S.; Mamone, G.; Ferranti, P. The performing protein: Beyond wheat proteomics? Cereal Chem. 2013, 90, 358–366. [Google Scholar] [CrossRef]
  15. Schalk, K.; Lexhaller, B.; Koehler, P.; Scherf, K.A. Isolation and characterization of gluten protein types from wheat, rye, barley and oats for use as reference materials. PLoS ONE 2017, 12, e0172819. [Google Scholar] [CrossRef]
  16. Rani, M.; Sogi, D.S.; Gill, B.S. Characterization of gliadin, secalin and hordein fractions using analytical techniques. Sci. Rep. 2021, 11, 23135. [Google Scholar] [CrossRef]
  17. Mickowska, B.; Socha, P.; Urminská, D. Immunochemical evaluation of proteolysis of cereal proteins causing celiac disease by microbial proteases. Food Agric. Immunol. 2016, 27, 743–757. [Google Scholar] [CrossRef]
  18. Lexhaller, B.; Colgrave, M.L.; Scherf, K.A. Characterization and relative quantitation of wheat, rye, and barley gluten protein types by liquid chromatography–tandem mass spectrometry. Front. Plant Sci. 2019, 10, 1530. [Google Scholar] [CrossRef] [PubMed]
  19. Cebolla, Á.; Moreno, M.D.L.; Coto, L.; Sousa, C. Gluten immunogenic peptides as standard for the evaluation of potential harmful prolamin content in food and human specimen. Nutrients 2018, 10, 1927. [Google Scholar] [CrossRef] [PubMed]
  20. Balakireva, A.V.; Zamyatnin, A.A., Jr. Properties of gluten intolerance: Gluten structure, evolution, pathogenicity and detoxification capabilities. Nutrients 2016, 8, 644. [Google Scholar] [CrossRef]
  21. Cabanillas, B. Gluten-related disorders: Celiac disease, wheat allergy, and nonceliac gluten sensitivity. Crit. Rev. Food Sci. Nutr. 2020, 60, 2606–2621. [Google Scholar] [CrossRef]
  22. Shan, L.; Molberg, Ø.; Parrot, I.; Hausch, F.; Filiz, F.; Gray, G.M.; Sollid, L.M.; Khosla, C. Structural basis for gluten intolerance in celiac sprue. Science 2002, 297, 2275–2279. [Google Scholar] [CrossRef]
  23. Ciccocioppo, R.; Di Sabatino, A.; Corazza, G.R. The immune recognition of gluten in coeliac disease. Clin. Exp. Immunol. 2005, 140, 408–416. [Google Scholar] [CrossRef]
  24. Camarca, A.; Del Mastro, A.; Gianfrani, C. Repertoire of gluten peptides active in celiac disease patients: Perspectives for translational therapeutic applications. Endocr. Metab. Immune Disord. Drug Targets 2012, 12, 207–219. [Google Scholar] [CrossRef]
  25. Sollid, L.M.; Tye-Din, J.A.; Qiao, S.W.; Anderson, R.P.; Gianfrani, C.; Koning, F. Update 2020: Nomenclature and listing of celiac disease-relevant gluten epitopes recognized by CD4+ T cells. Immunogenetics 2020, 72, 85–88. [Google Scholar] [CrossRef] [PubMed]
  26. Chlubnová, M.; Christophersen, A.O.; Sandve, G.K.F.; Lundin, K.E.A.; Jahnsen, J.; Dahal-Koirala, S.; Sollid, L.M. Identification of gluten T cell epitopes driving celiac disease. Sci. Adv. 2023, 9, eade5800. [Google Scholar] [CrossRef] [PubMed]
  27. Arentz-Hansen, H.; McAdam, S.N.; Molberg, Ø.; Fleckenstein, B.; Lundin, K.E.; Jørgensen, T.J.; Jung, G.; Roepstorff, P.; Sollid, L.M. Celiac lesion T cells recognize epitopes that cluster in regions of gliadins rich in proline residues. Gastroenterology 2002, 123, 803–809. [Google Scholar] [CrossRef]
  28. Sjöström, H.; Lundin, K.E.; Molberg, O.; Körner, R.; McAdam, S.N.; Anthonsen, D.; Quarsten, H.; Norén, O.; Roepstorff, P.; Thorsby, E.; et al. Identification of a gliadin Tcell epitope in coeliac disease: General importance of gliadin deamidation for intestinal T-cell recognition. Scand. J. Immunol. 1998, 48, 111–115. [Google Scholar] [CrossRef]
  29. Vader, W.; Kooy, Y.; Van Vellen, P.; De Ru, A.; Harris, D.; Benckhuijsen, W.; Pena, S.; Mearin, L.; Drijfhout, J.W.; Koning, F. The gluten response in children with celiac disease is directed toward multiple gliadin and glutenin peptides. Gastroenterology 2002, 122, 1729–1737. [Google Scholar] [CrossRef]
  30. Mamone, G.; Ferranti, P.; Melck, D.; Tafuro, F.; Longobardo, L.; Chianese, L.; Addeo, F. Susceptibility to transglutaminase of gliadin peptides predicted by a mass spectrometrybased assay. FEBS Lett. 2004, 562, 177–182. [Google Scholar] [CrossRef] [PubMed]
  31. Dørum, S.; Arntzen, M.Ø.; Qiao, S.W.; Holm, A.; Koehler, C.J.; Thiede, B.; Sollid, L.M.; Fleckenstein, B. The preferred substrates for transglutaminase 2 in a complex wheat gluten digest are Peptide fragments harboring celiac disease T-cell epitopes. PLoS ONE 2010, 19, e14056. [Google Scholar] [CrossRef]
  32. Vader, L.W.; Stepniak, D.T.; Bunnik, E.M.; Kooy, Y.M.; de Haan, W.; Drijfhout, J.W.; Van Veelen, P.A.; Koning, F. Characterization of cereal toxicity for celiac disease patients based on protein homology in grains. Gastroenterology 2003, 125, 1105–1113. [Google Scholar] [CrossRef]
  33. Tye-Din, J.A.; Stewart, J.A.; Dromey, J.A.; Beissbarth, T.; van Heel, D.A.; Tatham, A.; Henderson, K.; Mannering, S.I.; Gianfrani, C.; Jewell, D.P.; et al. Comprehensive, quantitative mapping of T cell epitopes in gluten in celiac disease. Sci. Transl. Med. 2010, 2, 41–51. [Google Scholar] [CrossRef]
  34. Gianfrani, C.; Camarca, A.; Mazzarella, G.; Di Stasio, L.; Giardullo, N.; Ferranti, P.; Picariello, G.; Rotondi Aufiero, V.; Picascia, S.; Troncone, R.; et al. Extensive in vitro gastrointestinal digestion markedly reduces the immune-toxicity of Triticum monococcum wheat: Implication for celiac disease. Mol. Nutr. Food Res. 2015, 59, 1844–1854. [Google Scholar] [CrossRef]
  35. Mamone, G.; Nitride, C.; Picariello, G.; Addeo, F.; Ferranti, P.; Mackie, A. Tracking the fate of pasta (T. durum semolina) immunogenic proteins by in vitro simulated digestion. J. Agric. Food Chem. 2015, 63, 2660–2667. [Google Scholar] [CrossRef] [PubMed]
  36. Boukid, F.; Prandi, B.; Faccini, A.; Sforza, S. A Complete Mass Spectrometry (MS)-Based Peptidomic Description of Gluten Peptides Generated During In Vitro Gastrointestinal Digestion of Durum Wheat: Implication for Celiac Disease. J. Am. Soc. Mass Spectrom. 2019, 30, 1481–1490. [Google Scholar] [CrossRef] [PubMed]
  37. Shan, L.; Qiao, S.W.; Arentz-Hansen, H.; Molberg, Ø.; Gray, G.M.; Sollid, L.M.; Khosla, C. Identification and analysis of multivalent proteolytically resistant peptides from gluten: Implications for celiac sprue. J. Proteome Res. 2005, 4, 1732–1741. [Google Scholar] [CrossRef]
  38. Barone, M.V.; Troncone, R.; Auricchio, S. Gliadin peptides as triggers of the proliferative and stress/innate immune response of the celiac small intestinal mucosa. Int. J. Mol. Sci. 2014, 15, 20518–20537. [Google Scholar] [CrossRef] [PubMed]
  39. Mamone, G.; Ferranti, P.; Rossi, M.; Roepstorff, P.; Fierro, O.; Malorni, A.; Addeo, F. Identification of a peptide from alpha-gliadin re-sistant to digestive enzymes: Implications for celiac disease. J. Chromatogr. B 2007, 855, 236–241. [Google Scholar] [CrossRef]
  40. Iacomino, G.; Fierro, O.; D’Auria, S.; Picariello, G.; Ferranti, P.; Liguori, C.; Addeo, F.; Mamone, G. Structural analysis and Caco-2 cell permeability of the celiac-toxic A-gliadin peptide 31–55. J. Agric. Food Chem. 2013, 61, 1088–1096. [Google Scholar] [CrossRef]
  41. Palanski, B.A.; Weng, N.; Zhang, L.; Hilmer, A.J.; Fall, L.A.; Swaminathan, K.; Jabri, B.; Sousa, C.; Fernandez-Becker, N.Q.; Khosla, C.; et al. An efficient urine peptidomics workflow identifies chemically defined dietary gluten peptides from patients with celiac disease. Nat. Commun. 2022, 13, 888. [Google Scholar] [CrossRef]
  42. Akobeng, A.K.; Singh, P.; Kumar, M.; Al Khodor, S. Role of the gut microbiota in the pathogenesis of coeliac disease and potential therapeutic implications. Eur. J. Nutr. 2020, 59, 3369–3390. [Google Scholar] [CrossRef]
  43. Wieser, H.; Segura, V.; Ruiz-Carnicer, Á.; Sousa, C.; Comino, I. Food safety and cross-contamination of gluten-free products: A narrative review. Nutrients 2021, 13, 2244. [Google Scholar] [CrossRef]
  44. Corazza, G.R.; Troncone, R.; Lenti, M.V.; Silano, M. Pediatric and Adult Celiac Disease: A Clinically Oriented Perspective; Elsevier: Amsterdam, The Netherlands, 2024. [Google Scholar]
  45. Rotondi Aufiero, V.; Fasano, A.; Mazzarella, G. Non-celiac gluten sensitivity: How its gut immune activation and potential dietary management differ from celiac disease. Mol. Nutr. Food Res. 2018, 62, 1700854. [Google Scholar] [CrossRef]
  46. Zevallos, V.F.; Raker, V.; Tenzer, S.; Jimenez-Calvente, C.; Ashfaq-Khan, M.; Rüssel, N.; Pickert, G.; Schild, H.; Steinbrink, K.; Schuppan, D. Nutritional Wheat Amylase-Trypsin Inhibitors Promote Intestinal Inflammation via Activation of Myeloid Cells. Gastroenterology 2017, 152, 1100–1113. [Google Scholar] [CrossRef] [PubMed]
  47. Vaquero, L.; Comino, I.; Vivas, S.; Rodríguez-Martín, L.; Giménez, M.J.; Pastor, J.; Sousa, C.; Barro, F. Tritordeum: A novel cereal for food processing with good acceptability and significant reduction in gluten immunogenic peptides in comparison with wheat. J. Sci. Food Agric. 2018, 98, 2201–2209. [Google Scholar] [CrossRef]
  48. De Caro, S.; Venezia, A.; Di Stasio, L.; Danzi, D.; Pignone, D.; Mamone, G.; Iacomino, G. Tritordeum: Promising Cultivars to Improve Health. Foods 2024, 13, 661. [Google Scholar] [CrossRef] [PubMed]
  49. Manza, F.; Lungaro, L.; Costanzini, A.; Caputo, F.; Carroccio, A.; Mansueto, P.; Seidita, A.; Raju, S.A.; Volta, U.; De Giorgio, R.; et al. Non-Celiac Gluten/Wheat Sensitivity-State of the Art: A Five-Year Narrative Review. Nutrients 2025, 17, 220. [Google Scholar] [CrossRef]
  50. Sapone, A.; Bai, J.C.; Ciacci, C.; Dolinsek, J.; Green, P.H.; Hadjivassiliou, M.; Kaukinen, K.; Rostami, K.; Sanders, D.S.; Schumann, M.; et al. Spectrum of gluten-related disorders: Consensus on new nomenclature and classification. BMC Med. 2012, 10, 13. [Google Scholar] [CrossRef] [PubMed]
  51. Tranquet, O.; Larré, C.; Denery-Papini, S. Allergic reactions to hydrolysed wheat proteins: Clinical aspects and molecular structures of the allergens involved. Crit. Rev. Food Sci. Nutr. 2020, 60, 147–156. [Google Scholar] [CrossRef]
  52. Denery-Papini, S.; Pineau, F.; Bodinier, M.; Branlard, G.; Faye, A.; Tranquet, O.; Larre, C.; Monoret-Vautrin, D.A. Allergens and epitopes involved in food allergy to wheat and their presence in different wheat genotypes. Gluten Proteins 2009, 280–284. [Google Scholar]
  53. Srisuwatchari, W.; Kanchanapoomi, K.; Pacharn, P. Molecular Diagnosis to IgE-mediated Wheat Allergy and Wheat-Dependent Exercise-Induced Anaphylaxis. Clin Rev Allergy Immunol. 2025, 68, 47. [Google Scholar] [CrossRef]
  54. Wang, Y.; Weng, J.; Zhu, C.; Ai, R.; Zhou, J.; Wang, C.; Chen, Q.; Fu, L. Allergenicity assessment and allergen profile analysis of different Chinese wheat cultivars. World Allergy Organ. J. 2021, 14, 100559. [Google Scholar] [CrossRef]
  55. Picariello, G.; Mamone, G.; Addeo, F.; Ferranti, P. The frontiers of mass spectrometry-based techniques in food allergenomics. J. Chromatogr. A 2011, 1218, 7386–7398. [Google Scholar] [CrossRef]
  56. Ricci, G.; Andreozzi, L.; Cipriani, F.; Giannetti, A.; Gallucci, M.; Caffarelli, C. Wheat allergy in children: A comprehensive update. Medicina 2019, 55, 400. [Google Scholar] [CrossRef]
  57. Ribeiro, M.; de Sousa, T.; Sabença, C.; Poeta, P.; Bagulho, A.S.; Igrejas, G. Advances in quantification and analysis of the celiac-related immunogenic potential of gluten. Compr. Rev. Food Sci. Food Saf. 2021, 20, 4278–4298. [Google Scholar] [CrossRef]
  58. Munshi, M.; Deb, S. Gluten Detection in Foods. In Challenges and Potential Solutions in Gluten Free Product Development; Springer International Publishing: Cham, Switzerland, 2021; pp. 111–140. [Google Scholar]
  59. Méndez, E.; Vela, C.; Immer, U.; Janssen, F.W. Report of a collaborative trial to investigate the performance of the R5 enzyme linked immunoassay to determine gliadin in gluten-free food. Eur. J. Gastroenterol. Hepatol. 2005, 17, 1053–1063. [Google Scholar] [CrossRef]
  60. Schopf, M.; Scherf, K.A. Wheat cultivar and species influence variability of gluten ELISA analyses based on polyclonal and monoclonal antibodies R5 and G12. J. Cereal Sci. 2018, 83, 32–41. [Google Scholar] [CrossRef]
  61. Panda, R.; Garber, E.A. Detection and quantitation of gluten in fermented-hydrolyzed foods by antibody-based methods: Challenges, progress, and a potential path forward. Front. Nutr. 2019, 6, 97. [Google Scholar] [CrossRef] [PubMed]
  62. Masiri, J.; Benoit, L.; Katepalli, M.; Meshgi, M.; Cox, D.; Nadala, C.; Sung, S.-L.; Samadpour, M. Novel monoclonal antibody-based immunodiagnostic assay for rapid detection of deamidated gluten residues. J. Agric. Food Chem. 2016, 64, 3678–3687. [Google Scholar] [CrossRef] [PubMed]
  63. Panda, R.; Boyer, M.; Garber, E.A. A multiplex competitive ELISA for the detection and characterization of gluten in fermented-hydrolyzed foods. Anal. Bioanal. Chem. 2017, 409, 6959–6973. [Google Scholar] [CrossRef]
  64. Comino, I.; Real, A.; Vivas, S.; Síglez, M.Á.; Caminero, A.; Nistal, E.; Casqueiro, J.; Rodríguez-Herrera, A.; Cebolla, A.; Sousa, C. Monitoring of gluten-free diet compliance in celiac patients by assessment of gliadin 33-mer equivalent epitopes in feces. Am. J. Clin. Nutr. 2012, 95, 670–677. [Google Scholar] [CrossRef]
  65. Scherf, K.A.; Poms, R.E. Recent developments in analytical methods for tracing gluten. J. Cereal Sci. 2016, 67, 112–122. [Google Scholar] [CrossRef]
  66. Segura, V.; Siglez, M.Á.; Ruiz-Carnicer, Á.; Martín-Cabrejas, I.; van der Hofstadt, M.; Mellado, E.; Comino, I.; Sousa, C. A highly sensitive method for the detection of hydrolyzed gluten in beer samples using LFIA. Foods 2022, 12, 160. [Google Scholar] [CrossRef] [PubMed]
  67. Slot, I.B.; Van Der Fels-Klerx, H.J.; Bremer, M.G.; Hamer, R.J. Immunochemical detection methods for gluten in food products: Where do we go from here? Crit. Rev. Food Sci. Nutr. 2016, 56, 2455–2466. [Google Scholar] [CrossRef]
  68. Alves, T.O.; D’Almeida, C.T.; Scherf, K.A.; Ferreira, M.S. Modern approaches in the identification and quantification of immunogenic peptides in cereals by LC-MS/MS. Front. Plant Sci. 2019, 10, 1470. [Google Scholar] [CrossRef]
  69. Lu, Y.; Ji, H.; Chen, Y.; Li, Z.; Timira, V. A systematic review on the recent advances of wheat allergen detection by mass spectrometry: Future prospects. Crit. Rev. Food Sci. Nutr. 2023, 63, 12324–12340. [Google Scholar] [CrossRef]
  70. Schalk, K.; Lang, C.; Wieser, H.; Koehler, P.; Scherf, K.A. Quantitation of the immunodominant 33-mer peptide from α-gliadin in wheat flours by liquid chromatography tandem mass spectrometry. Sci. Rep. 2017, 7, 45092. [Google Scholar] [CrossRef]
  71. Malalgoda, M.; Meinhardt, S.W.; Simsek, S. Detection and quantitation of immunogenic epitopes related to celiac disease in historical and modern hard red spring wheat cultivars. Food Chem. 2018, 264, 101–107. [Google Scholar] [CrossRef]
  72. de Lourdes Moreno, M.; Cebolla, Á.; Muñoz-Suano, A.; Carrillo-Carrion, C.; Comino, I.; Pizarro, Á.; León, F.; Rodríguez-Herrera, A.; Sousa, C. Detection of gluten immunogenic peptides in the urine of patients with coeliac disease reveals transgressions in the gluten-free diet and incomplete mucosal healing. Gut 2017, 66, 250–257. [Google Scholar] [CrossRef]
  73. Murray, J.A.; Syage, J.A.; Wu, T.T.; Dickason, M.A.; Ramos, A.G.; Van Dyke, C.; Horwath, I.; Lavin, P.T.; Mäki, M.; Hujoel, I.; et al. CeliacShield Study Group. Latiglutenase Protects the Mucosa and Attenuates Symptom Severity in Patients with Celiac Disease Exposed to a Gluten Challenge. Gastroenterology 2022, 163, 1510–1521. [Google Scholar] [CrossRef] [PubMed]
  74. Weng, Z.; Chen, Y.; Liang, T.; Lin, Y.; Cao, H.; Song, H.; Xiong, L.; Wang, F.; Shen, X.; Xiao, J. A review on processing methods and functions of wheat germ-derived bioactive peptides. Crit. Rev. Food Sci. Nutr. 2023, 63, 5577–5593. [Google Scholar] [CrossRef]
  75. Karami, Z.; Butkinaree, C.; Somsong, P.; Duangmal, K. Assessment of the DPP-IV inhibitory potential of mung bean and adzuki bean protein hydrolysates using enzymatic hydrolysis process: Specificity of peptidases and novel peptides. Int. J. Food Sci. Technol. 2023, 58, 2995–3005. [Google Scholar] [CrossRef]
  76. Mora, L.; Toldrá, F. Advanced enzymatic hydrolysis of food proteins for the production of bioactive peptides. Curr. Opin. Food Sci. 2023, 49, 1009. [Google Scholar] [CrossRef]
  77. Zhang, Z.H.; Cheng, W.L.; Li, X.D.; Wang, X.; Yang, F.W.; Xiao, J.S.; Li, Y.X.; Zhao, G.P. Extraction, bioactive function and application of wheat germ protein/peptides: A review. Curr. Res. Food Sci. 2023, 6, 100512. [Google Scholar] [CrossRef]
  78. Liu, W.; Ren, J.; Wu, H.; Zhang, X.; Han, L.; Gu, R. Inhibitory effects and action mechanism of five antioxidant peptides derived from wheat gluten on cells oxidative stress injury. Food Biosci. 2023, 56, 103236. [Google Scholar] [CrossRef]
  79. Li-Chan, E.C.Y. Bioactive peptides and protein hydrolysates: Research trends and challenges for application as nutraceuticals and functional food ingredients. Curr. Opin. Food Sci. 2015, 1, 28–37. [Google Scholar] [CrossRef]
  80. Liu, W.Y.; Zhang, J.T.; Miyakawa, T.; Li, G.M.; Gu, R.Z.; Tanokura, M. Antioxidant properties and inhibition of angiotensin-converting enzyme by highly active peptides from wheat gluten. Sci. Rep. 2021, 11, 5206. [Google Scholar] [CrossRef]
  81. Asoodeh, A.; Haghighi, L.; Chamani, J.; Ansari-Ogholbeyk, M.A.; Mojallal-Tabatabaei, Z.; Lagzian, M. Potential angiotensin I converting enzyme inhibitory peptides from gluten hydrolysate: Biochemical characterization and molecular docking study. J. Cereal Sci. 2014, 60, 6092–6098. [Google Scholar] [CrossRef]
  82. Wang, X.; Fu, J.; Bhullar, K.S.; Chen, B.; Liu, H.; Zhang, Y.; Wang, C.; Liu, C.; Su, D.; Ma, X.; et al. Identification, in silico selection, and mechanistic investigation of antioxidant peptides from corn gluten meal hydrolysate. Food Chem. 2024, 446, 138777. [Google Scholar] [CrossRef] [PubMed]
  83. Kaur, L.; Mao, B.; Beniwal, A.S.; Abhilasha, R.; Kaur, F.M.; Singh Chian, J. Alternative proteins vs animal proteins: The influence of structure and processing on their gastro-small intestinal digestion. Trends Food Sci. Technol. 2022, 122, 275–286. [Google Scholar] [CrossRef]
  84. Manai, F.; Zanoletti, L.; Morra, G.; Mansoor, S.; Carriero, F.; Bozzola, E.; Muscianisi, S.; Comincini, S. Gluten Exorphins Promote Cell Proliferation through the Activation of Mitogenic and Pro-Survival Pathways. Int. J. Mol. Sci. 2023, 24, 3912. [Google Scholar] [CrossRef]
  85. Croall, I.D.; Hoggard, N.; Hadjivassiliou, M. Gluten and autism spectrum disorder. Nutrients 2021, 13, 572. [Google Scholar] [CrossRef] [PubMed]
  86. Di Liberto, D.; D’Anneo, A.; Carlisi, D.; Emanuele, S.; De Blasio, A.; Calvaruso, G.; Giuliano, M.; Lauricella, M. Brain opioid activity and oxidative injury: Different molecular scenarios connecting celiac disease and autistic spectrum disorder. Brain Sci. 2020, 10, 437. [Google Scholar] [CrossRef] [PubMed]
  87. Kaur, J.; Kumar, V.; Sharma, K.; Kaur, S.; Gat, Y.; Goyal, A.; Tanwar, B. Opioid peptides: An overview of functional significance. Int. J. Pept. Res. Ther. 2020, 26, 33–41. [Google Scholar] [CrossRef]
  88. Takahashi, M.; Fukunaga, H.; Kaneto, H.; Fukudome, S.; Yoshikawa, M. Behavioral and pharmacological studies on gluten exorphin A5, a newly isolated bioactive food protein fragment, in mice. Jpn. J. Pharmacol. 2000, 84, 259–265. [Google Scholar] [CrossRef]
  89. Ergün, C.; Urhan, M.; Ayer, A. A review on the relationship between gluten and schizophrenia: Is gluten the cause? Nutr. Neurosci. 2018, 21, 455–466. [Google Scholar] [CrossRef]
  90. Samaroo, D.; Dickerson, F.; Kasarda, D.D.; Green, P.H.; Briani, C.; Yolken, R.H.; Alaedini, A. Novel immune response to gluten in individuals with schizophrenia. Schizophr. Res. 2010, 118, 248–255. [Google Scholar] [CrossRef]
  91. Cruz-Chamorro, I.; Álvarez-Sánchez, N.; Santos-Sánchez, G.; Pedroche, J.; Fernández-Pachón, M.S.; Millán, F.; Millán-Linares, M.C.; Lardone, P.J.; Bejarano, I.; Guerrero, J.; et al. Immunomodulatory and Antioxidant Properties of Wheat Gluten Protein Hydrolysates in Human Peripheral Blood Mononuclear Cells. Nutrients 2020, 12, 1673. [Google Scholar] [CrossRef]
  92. Mamone, G.; Picariello, G. Optimized extraction and large-scale proteomics of pig jejunum brush border membranes for use in in vitro digestion models. Food Res. Int. 2023, 164, 112326. [Google Scholar] [CrossRef]
  93. Di Stasio, L.; De Caro, S.; Marulo, S.; Ferranti, P.; Picariello, G.; Mamone, G. Impact of porcine brush border membrane enzymes on INFOGEST in vitro digestion model: A step forward to mimic the small intestinal phase. Food Res. Int. 2024, 197, 115300. [Google Scholar] [CrossRef]
  94. Singh, P.; Arora, A.; Strand, T.A.; Leffler, D.A.; Catassi, C.; Green, P.H.; Kelly, C.P.; Ahuja, V.; Makharia, G.K. Global Prevalence of Celiac Disease: Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2018, 16, 823–836. [Google Scholar] [CrossRef]
  95. Santonicola, A.; Soldaini, C.; Ciacci, C. New therapies in celiac disease. Curr. Opin. Gastroenterol. 2025, 41, 124–131. [Google Scholar] [CrossRef]
  96. Caputo, I.; Lepretti, M.; Martucciello, S.; Esposito, C. Enzymatic strategies to detoxify gluten: Implications for celiac disease. Enzym. Res. 2010, 174354. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, Y.; Wu, H.; Fu, L. A review of gluten detoxification in wheat for food applications: Approaches, mechanisms, and implications. Crit. Rev. Food Sci. Nutr. 2025, 65, 2100–2116. [Google Scholar] [CrossRef] [PubMed]
  98. Møller, M.S.; Svensson, B. Enzymes in grain processing. Curr. Opin. Food Sci. 2021, 37, 153–159. [Google Scholar] [CrossRef]
  99. Sakandar, H.A.; Usman, K.; Imran, M. Isolation and characterization of gluten-degrading Enterococcus mundtii and Wickerhamomyces anomalus, potential probiotic strains from indigenously fermented sourdough (Khamir). LWT 2018, 91, 271–277. [Google Scholar] [CrossRef]
  100. Reale, A.; Di Stasio, L.; Di Renzo, T.; De Caro, S.; Ferranti, P.; Picariello, G.; Addeo, F.; Mamone, G. Bacteria do it better! Proteomics suggests the molecular basis for improved digestibility of sourdough products. Food Chem. 2021, 359, 129955. [Google Scholar] [CrossRef]
  101. Bradauskiene, V.; Vaiciulyte-Funk, L.; Shah, B.R.; Cernauskas, D.; Tita, M.A. Recent advances in biotechnological methods for wheat gluten immunotoxicity abolishment–a review. Pol. J. Food Nutr. Sci. 2021, 71, 5–20. [Google Scholar] [CrossRef]
  102. Socha, P.; Mickowska, B.; Urminská, D.; Kačmárová, K. The use of different proteases to hydrolyze gliadins. J. Microbiol. Biotechnol. Food Sci. 2015, 4, 101–104. [Google Scholar] [CrossRef]
  103. de la Barca, A.C.; Heredia-Sandoval, N.G.; Mejía-León, M.E. Detoxification of wheat proteins by enzymatic technology. In Biotechnological Strategies for the Treatment of Gluten Intolerance; Academic Press: Cambridge, MA, USA, 2021; pp. 155–176. [Google Scholar]
  104. Diaz-Mendoza, M.; Diaz, I.; Martinez, M. Insights on the proteases involved in barley and wheat grain germination. Int. J. Mol. Sci. 2019, 20, 2087. [Google Scholar] [CrossRef]
  105. Bradauskiene, V.; Vaiciulyte-Funk, L.; Cernauskas, D.; Dzingeleviciene, R.; Lima, J.P.; Bradauskaite, A.; Tita, M.A. The efficacy of plant enzymes bromelain and papain as a tool for reducing gluten immunogenicity from wheat bran. Processes 2022, 10, 1948. [Google Scholar] [CrossRef]
  106. David Troncoso, F.; Alberto Sánchez, D.; Luján Ferreira, M. Production of plant proteases and new biotechnological applications: An updated review. ChemistryOpen 2022, 11, e202200017. [Google Scholar] [CrossRef]
  107. Li, Y.; Yu, J.; Goktepe, I.; Ahmedna, M. The potential of papain and alcalase enzymes and process optimizations to reduce allergenic gliadins in wheat flour. Food Chem. 2016, 196, 1338–1345. [Google Scholar] [CrossRef]
  108. Song, P.; Zhang, X.; Wang, S.; Xu, W.; Wang, F.; Fu, R.; Wei, F. Microbial proteases and their applications. Front. Microbiol. 2023, 14, 1236368. [Google Scholar] [CrossRef]
  109. Rossi, S.; Giordano, D.; Mazzeo, M.F.; Maurano, F.; Luongo, D.; Facchiano, A.; Siciliano, R.A.; Rossi, M. Transamidation down-regulates intestinal immunity of recombinant α-gliadin in HLA-DQ8 transgenic mice. Int. J. Mol. Sci. 2021, 22, 7019. [Google Scholar] [CrossRef]
  110. Treppiccione, L.; Picarelli, A.; Rossi, M. Beneficial role of microbial transglutaminase in the pathogenetic mechanisms of coeliac disease. J. Pediatr. Gastroenterol. Nutr. 2022, 74, 728–733. [Google Scholar] [CrossRef]
  111. Yoosuf, S.; Makharia, G.K. Evolving therapy for celiac disease. Front. Pediatr. 2019, 7, 193. [Google Scholar] [CrossRef]
  112. Alvarez-Sieiro, P.; Martin, M.C.; Redruello, B.; Del Rio, B.; Ladero, V.; Palanski, B.A.; Khosla, C.; Fernandez, M.; Alvarez, M.A. Generation of food-grade recombinant Lactobacillus casei delivering Myxococcus xanthus prolyl endopeptidase. Appl. Microbiol. Biotechnol. 2014, 98, 6689–6700. [Google Scholar] [CrossRef] [PubMed]
  113. Álvarez Sieiro, P.; Álvarez Sieiro, M.C.; Redruello, B.; Ladero Losada, V.M.; Fernández García, M.; Álvarez González, M.Á. New Strain of Lactobacillus Casei with Ability to Degrade the Immunotoxic Peptide from Gluten. U.S. Patent 20170196918A1, 22 May 2015. [Google Scholar]
  114. Savvateeva, L.V.; Chepikova, O.E.; Solonkina, A.D.; Sakharov, A.A.; Gorokhovets, N.V.; Golovin, A.V.; Zamyatnin, A.A., Jr. Computational Screening and Experimental Evaluation of Wheat Proteases for Use in the Enzymatic Therapy of Gluten-Related Disorders. Pharmaceuticals 2025, 18, 592. [Google Scholar] [CrossRef]
  115. Savvateeva, L.V.; Gorokhovets, N.V.; Makarov, V.A.; Serebryakova, M.V.; Solovyev, A.G.; Morozov, S.Y.; Reddy, V.P.; Zernii, E.Y.; Zamyatnin, A.A.; Aliev, G. Glutenase and collagenase activities of wheat cysteine protease Triticain-α: Feasibility for enzymatic therapy assays. Int. J. Biochem. Cell Biol. 2015, 62, 115–124. [Google Scholar] [CrossRef] [PubMed]
  116. Cavaletti, L.; Taravella, A.; Carrano, L.; Carenzi, G.; Sigurtà, A.; Solinas, N.; De Caro, S.; Di Stasio, L.; Picascia, S.; Laezza, M.; et al. E40, a novel microbial protease efficiently detoxifying gluten proteins, for the dietary management of gluten intolerance. Sci. Rep. 2019, 9, 13147. [Google Scholar] [CrossRef]
  117. Mamone, G.; Comelli, M.C.; Vitale, S.; Di Stasio, L.; Kessler, K.; Mottola, I.; Siano, F.; Cavaletti, L.; Gianfrani, C. E40 glutenase detoxification capabilities of residual gluten immunogenic peptides in in vitro gastrointestinal digesta of food matrices made of soft and durum wheat. Front. Nutr. 2022, 9, 974771. [Google Scholar] [CrossRef] [PubMed]
  118. Wolf, C.; Siegel, J.B.; Tinberg, C.; Camarca, A.; Gianfrani, C.; Paski, S.; Guan, R.; Montelione, G.; Baker, D.; Pultz, I.S. Engineering of Kuma030: A gliadin peptidase that rapidly degrades immunogenic gliadin peptides in gastric conditions. J. Am. Chem. Soc. 2015, 137, 13106–13113. [Google Scholar] [CrossRef]
  119. Pultz, I.S.; Hill, M.; Vitanza, J.M.; Wolf, C.; Saaby, L.; Liu, T.; Winkle, P.; Leffler, D.A. Gluten degradation, pharmacokinetics, safety, and tolerability of TAK-062, an engineered enzyme to treat celiac disease. Gastroenterology 2021, 161, 81–93. [Google Scholar] [CrossRef] [PubMed]
  120. Picascia, S.; Camarca, A.; Malamisura, M.; Mandile, R.; Galatola, M.; Cielo, D.; Gazza, L.; Mamone, G.; Auricchio, S.; Troncone, R.; et al. In Celiac Disease Patients the In Vivo Challenge with the Diploid Triticum monococcum Elicits a Reduced Immune Response Compared to Hexaploid Wheat. Mol. Nutr. Food Res. 2020, 64, e1901032. [Google Scholar] [CrossRef] [PubMed]
  121. Kalunke, R.M.; Tundo, S.; Sestili, F.; Camerlengo, F.; Lafiandra, D.; Lupi, R.; Larré, C.; Denery-Papini, S.; Islam, S.; Ma, W.; et al. Reduction of allergenic potential in bread wheat RNAi transgenic lines silenced for CM3, CM16 and 0.28 ATI genes. Int. J. Mol. Sci. 2020, 21, 5817. [Google Scholar] [CrossRef]
  122. Jouanin, A.; Gilissen, L.J.; Schaart, J.G.; Leigh, F.J.; Cockram, J.; Wallington, E.J.; Boyd, L.A.; van den Broeck, H.C.; Van Der Meer, I.M.; America, A.H.P.; et al. CRISPR/Cas9 gene editing of gluten in wheat to reduce gluten content and exposure—Reviewing methods to screen for coeliac safety. Front. Nutr. 2020, 7, 51. [Google Scholar] [CrossRef]
  123. Gupta, A.; Hua, L.; Zhang, Z.; Yang, B.; Li, W. CRISPR-induced miRNA156-recognition element mutations in TaSPL13 improve multiple agronomic traits in wheat. Plant Biotechnol. J. 2023, 21, 536–548. [Google Scholar] [CrossRef]
  124. Sánchez-León, S.; Marín-Sanz, M.; Guzmán-López, M.H.; Gavilán-Camacho, M.; Simón, E.; Barro, F. CRISPR/Cas9-mediated multiplex gene editing of gamma and omega gliadins: Paving the way for gliadin-free wheat. J. Exp. Bot. 2024, 75, 7079–7095. [Google Scholar] [CrossRef]
  125. Jouanin, A.; Gilissen, L.J.; Boyd, L.A.; Cockram, J.; Leigh, F.J.; Wallington, E.J.; van den Broeck, H.C.; van der Meer, I.M.; Schaart, J.G.; Visser, R.G.F.; et al. Food processing and breeding strategies for coeliac-safe and healthy wheat products. Food Res. Int. 2018, 110, 11–21. [Google Scholar] [CrossRef]
  126. Kim, S.; Sim, J.R.; Gu, Y.Q.; Altenbach, S.B.; Denery-Papini, S.; Pineau, F.; Tranquet, O.; Yang, Y.-J.; Park, E.J.; Lim, S.-H.; et al. Toward reducing the immunogenic potential of wheat flour: Identification and characterization of wheat lines missing omega-5 gliadins encoded by the 1D chromosome. Theor. Appl. Genet. 2023, 136, 33. [Google Scholar] [CrossRef]
  127. Nye-Wood, M.G.; Juhász, A.; Bose, U.; Colgrave, M.L. Proteome analysis and epitope mapping in a commercial reduced-gluten wheat product. Front. Nutr. 2021, 8, 705822. [Google Scholar] [CrossRef]
  128. Yang, J.; Pan, M.; Han, R.; Yang, X.; Liu, X.; Yuan, S.; Wang, S. Food irradiation: An emerging processing technology to improve the quality and safety of foods. Food Rev. Int. 2024, 40, 2321–2343. [Google Scholar] [CrossRef]
  129. Sakr, A.M. Evaluation of wheat germination for making gluten-free bread. Curr. Sci. Int. 2020, 9, 677–689. [Google Scholar]
  130. Adrianos, S.L.; Mattioni, B.; Tilley, M. Confirmation of gluten-free status of wheatgrass (Triticum aestivum). Qual. Assur. Saf. Crops Foods 2017, 9, 123–128. [Google Scholar] [CrossRef]
  131. Di Cagno, R.; De Angelis, M.; Lavermicocca, P.; De Vincenzi, M.; Giovannini, C.; Faccia, M.; Gobbetti, M. Proteolysis by sourdough lactic acid bacteria: Effects on wheat flour protein fractions and gliadin peptides involved in human cereal intolerance. Appl. Environ. Microbiol. 2002, 68, 623–633. [Google Scholar] [CrossRef]
  132. Di Cagno, R.; De Angelis, M.; Auricchio, S.; Greco, L.; Clarke, C.; De Vincenzi, M.; Giovannini, C.; D’Archivio, M.; Landolfo, F.; Parrilli, G.; et al. Sourdough bread made from wheat and nontoxic flours and started with selected lactobacilli is tolerated in celiac sprue patients. Appl. Environ. Microbiol. 2004, 70, 1088–1096. [Google Scholar] [CrossRef]
  133. Fernández-Peláez, J.; Paesani, C.; Gómez, M. Sourdough technology as a tool for the development of healthier grain-based products: An update. Agronomy 2020, 10, 1962. [Google Scholar] [CrossRef]
  134. De Almeida, N.E.C.; Esteves, F.G.; dos Santos-Pinto, J.R.A.; Peres de Paula, C.; Da Cunha, A.F.; Malavazi, I.; Palma, M.S.; Rodrigues-Filho, E. Digestion of intact gluten proteins by Bifidobacterium species: Reduction of cytotoxicity and proinflammatory responses. J. Agric. Food Chem. 2020, 68, 4485–4492. [Google Scholar] [CrossRef] [PubMed]
  135. Yin, Y.; Wang, J.; Yang, S.; Feng, J.; Jia, F.; Zhang, C. Protein degradation in wheat sourdough fermentation with Lactobacillus plantarum M616. Interdiscip. Sci. Comput. Life Sci. 2015, 7, 205–210. [Google Scholar] [CrossRef] [PubMed]
  136. El Mecherfi, K.E.; Lupi, R.; Cherkaoui, M.; Albuquerque, M.A.; Todorov, S.D.; Tranquet, O.; Klingebiel, C.; Rogniaux, H.; Denery-Papini, S.; Onno, B.; et al. Fermentation of gluten by Lactococcus lactis LLGKC18 reduces its antigenicity and allergenicity. Probiotics Antimicrob. Proteins 2022, 14, 779–791. [Google Scholar] [CrossRef]
  137. Leszczyńska, J.; Szczepankowska, A.K.; Majak, I.; Mańkowska, D.; Smolińska, B.; Ścieszka, S.; Diowksz, A.; Cukrowska, B.; Aleksandrzak-Piekarczyk, T. Reducing immunoreactivity of gluten peptides by probiotic lactic acid bacteria for dietary management of gluten-related diseases. Nutrients 2024, 16, 976. [Google Scholar] [CrossRef]
  138. Zadeike, D.; Cipkute, K.; Cizeikiene, D. Effects of Co-Fermentation with Lactic Acid Bacteria and Yeast on Gliadin Degradation in Whole-Wheat Sourdough. Fermentation 2025, 11, 238. [Google Scholar] [CrossRef]
  139. Malalgoda, M.; Manthey, F.; Simsek, S. Reducing the celiac disease antigenicity of wheat. Cereal Chem. 2018, 95, 49–58. [Google Scholar] [CrossRef]
  140. Iacomino, G.; Rotondi Aufiero, V.; Di Stasio, L.; Picascia, S.; Iannaccone, N.; Giardullo, N.; Troncone, R.; Gianfrani, C.; Mamone, G.; Mazzarella, G. Triticum monococcum amylase trypsin inhibitors possess a reduced potential to elicit innate immune response in celiac patients compared to Triticum aestivum. Food Res. Int. 2021, 145, 110386. [Google Scholar] [CrossRef] [PubMed]
  141. Mamone, G.; Iacomino, G. Comparison of the in vitro toxicity of ancient Triticum monococcum varieties ID331 and Monlis. Int. J. Food Sci. Nutr. 2018, 69, 954–962. [Google Scholar] [CrossRef]
  142. Di Stasio, L.; Picascia, S.; Auricchio, R.; Vitale, S.; Gazza, L.; Picariello, G.; Gianfrani, C.; Mamone, G. Comparative analysis of in vitro digestibility and immunogenicity of gliadin proteins from durum and einkorn wheat. Front. Nutr. 2020, 7, 56. [Google Scholar] [CrossRef] [PubMed]
  143. Graça, C.; Lima, A.; Raymundo, A.; Sousa, I. Sourdough fermentation as a tool to improve the nutritional and health-promoting properties of its derived-products. Fermentation 2021, 7, 246. [Google Scholar] [CrossRef]
  144. Stefańska, I.; Piasecka-Jóźwiak, K.; Kotyrba, D.; Kolenda, M.; Stecka, K.M. Selection of lactic acid bacteria strains for the hydrolysis of allergenic proteins of wheat flour. J. Sci. Food Agric. 2016, 96, 3897–3905. [Google Scholar] [CrossRef]
  145. Tsafrakidou, P.; Michaelidou, A.M.; Biliaderis, C.G. Fermented cereal-based products: Nutritional aspects, possible impact on gut microbiota and health implications. Foods 2020, 9, 734. [Google Scholar] [CrossRef] [PubMed]
  146. Fraberger, V.; Ladurner, M.; Nemec, A.; Grunwald-Gruber, C.; Call, L.M.; Hochegger, R.; Domig, K.J.; D’Amico, S. Insights into the potential of sourdough-related lactic acid bacteria to degrade proteins in wheat. Microorganisms 2020, 8, 1689. [Google Scholar] [CrossRef] [PubMed]
  147. Jaglan, A.; Sadera, G.; Goel, G. Proteolytic efficiency and degradation of gliadin and toxic immunopeptide in laboratory media by indigenous potential probiotic Bacillus strains. Food Humanit. 2025, 4, 100567. [Google Scholar] [CrossRef]
  148. Thiele, C.; Grassl, S.; Gänzle, M. Gluten hydrolysis and depolymerization during sourdough fermentation. J. Agric. Food Chem. 2004, 52, 1307–1314. [Google Scholar] [CrossRef]
  149. Nikoloudaki, O.; Celano, G.; Polo, A.; Cappello, C.; Granehäll, L.; Costantini, A.; Vacca, M.; Speckmann, B.; Di Cagno, R.; Francavilla, R.; et al. Novel probiotic preparation with in vivo gluten-degrading activity and potential modulatory effects on the gut microbiota. Microbiol. Spectr. 2024, 12, e03524-23. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Potential biological activities of wheat-derived bioactive peptides released by enzymatic hydrolysis. The figure was created with BioRender: https://www.biorender.com/ (accessed on 2 September 2025).
Figure 1. Potential biological activities of wheat-derived bioactive peptides released by enzymatic hydrolysis. The figure was created with BioRender: https://www.biorender.com/ (accessed on 2 September 2025).
Foods 14 03403 g001
Figure 2. Biological and genetic strategies for the detoxification of wheat-based products. References [37,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128]. Adapted and retrieved from BioRender templates: BioRender: https://www.biorender.com/ (accessed on 11 September 2025).
Figure 2. Biological and genetic strategies for the detoxification of wheat-based products. References [37,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128]. Adapted and retrieved from BioRender templates: BioRender: https://www.biorender.com/ (accessed on 11 September 2025).
Foods 14 03403 g002
Table 1. Overview of enzymatic, microbial, genetic, and oral therapeutic strategies for gluten detoxification.
Table 1. Overview of enzymatic, microbial, genetic, and oral therapeutic strategies for gluten detoxification.
Strategy TypeProcess or Method/Enzyme/Microbial Strain/Gene/CompoundExperimental Conditions/
Application Method
Mechanism of Action/Impact on GlutenReferences
Enzymatic
degradation
GerminationEndogenous enzymes of wheat grains8 days (25 °C, 100% humidity)Reduction of peptides eliciting immune response.[101,129]
Endogenous enzymes of wheat grains7 days (20 °C, pH 5.5, 100% humidity)Degradation of gluten (<LOD).[130]
Cysteine protease from barley germination (EP-B2)36 h, 100% humidityHydrolysis of immunotoxic sequences.[104]
Microbial
fermentation
Fermentation with LABLactobacillus species: alimentarius 15M, brevis 14G, sanfranciscensis 7A, plantarum CF1, and hilgardii 51B24 h at 37 °CGluten degradation.[131,132]
Streptococcus thermophilus, Lactobacillus. plantarum, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus delbrueckii subsp. Bulgaricus72 h at 30 °C under anaerobic conditionsGluten degradation.[101,133]
Bifidobacterium species: bifidum, longum, breve, animalis and infantis24 h at 30 °C under anaerobic conditionsProteolysis of gluten proteins.[134]
Lactobacillus. plantarum species24 h at 30 °CHydrolysis and solubilization of wheat proteins.[135]
Lactobacillus. sanfranciscensis24 h at 30 °CGluten degradation.[102]
Lactococcus lactis LLGKC1824 h at 37 °CDegradation of immunodominant sequences of ω-5 gliadin[136]
Combination of Lactobacillus casei LC130 and Lactobacillus paracasei LPC100 with the Streptococcus thermophilus2 h at 37 °CHydrolyzation of immunoreactive gliadin peptides.[137]
Combination of Lactobacillus. brevis and Pediococcus pentosaceus24 h at 35 °CReduced toxic gluten peptides content.[138]
Bacillus species: stearothermophilus, subtilis, tequilensis, licheniformis, thermoproteolyticus and cereus QAUSD071 h at 70 °C pH 8 or 24 h at 37 °CDegradation of immunogenic peptides such as the 33-mer.[102,134]
Fermentation with endo- and exoprotease/peptidase from bacteria, fungi, plants and animalsProline endoproteases (PEP) from Flavobacterium meningosepticum, Sphingomonas capsulata, and Myxococcus xanthus24 h at 37 °CReduction of immunoreactive gliadin peptides.[112,113,136]
Alcalase1 h at 50 °CReduced the antigenicity of wheat gluten hydrolysates.[107]
Plants protease from Nigella Sativa, Nepenthes pitcher (Neprosis), Carica papaya latex (Caricain)2 h at 37 °C; 80 min at 37 °CHydrolysis of gluten proteins.[106]
Plants protease as papain, bromelain and actinidin1 h at 65 °C or 70 °CHydrolysis of gluten proteins.[105,106,107]
TransamidationMicrobial transglutaminase (mTG)mTG catalyses the transamidation reaction that leads to the formation of an isopeptide bond.Prevention of the formation of immunodominant gluten peptides.[109,110]
Oral therapyEnzyme supplement therapy using either bacterial or fungal endopeptidases or endoproteasesProline endoproteases (PEP) from Flavobacterium meningosepticum, Sphingomonas capsulata, and Myxococcus xanthus.Optimal activity pH range of 3–5 (gastric phase)Efficiently degrades the most immune-toxic gluten.[37]
Kuma 030: engineered endopeptidase derived from the microbial protease of Alicyclobacillus sendaiensis (kumamolisin)Optimal activity pH 4 (gastric phase)Efficiently degrades the most immune-toxic gluten.[118]
TAK-062: engineered endopeptidase from the precursor Kuma030Optimal activity pH 2.5–6.0 (both gastric and intestinal phase)Degrade > 99% of gluten in a complex study meal in the human stomach and in vitro.[119]
Aspergillopepsin (ASP) from Aspergirllus nigerOptimal activity pH 3 (gastric phase)Hydrolyzation of immunoreactive gliadin peptides. Often used in combination with as EP-B2 or microbial prolyl endopeptidases to enhance the therapeutic efficacy.[117]
DPP-IV from Aspergillus oryzaeOptimal activity pH 7 (intestinal phase)Limited proteolytic effect alone. Often used in combination with PEP.[75]
Cysteine protease from wheat germination (Triticain-α)Optimal activity pH 3–6 (gastric phase)Hydrolysis of immunotoxic sequences of α-, γ-, ω-gliadin, and glutenin.[115]
E40 glutenase from Actinomycete strain Actinoallomurus A8Optimal activity pH range of 3–6 (gastric phase)Degradation of the immunodominant 33-mer peptide and whole gliadin proteins.[116,117]
Genetic
modification
CRISPR/Cas9Mutations of Gli-γ1-1D, Gli-γ2-1B,γ-gliadins, α-gliadinsGene-editing tool that uses a special protein (Cas9) to cut DNA at exact spots in order to add, remove, or change specific genes.Removing or reducing the toxic fractions of gluten.[122,123,124]
Chinese SpringDeletion of the 6DS armRemove specific chromosomal regionsStrong reduction of gliadin immunogenicity.[126]
Gamma irradiationDeletions in gliadins genesIt cause DNA strand breaks, which may lead to genetic alterations such as deletions, substitutions, or insertions.Removing or reducing the toxic fractions of gluten.[128]
RNAiTranslation in gliadins genesIt cause target mRNA degradation, which may lead to genetic alterations Reducing gliadin expression and developing wheat lines with fewer gluten genes and/or gluten genes with inactivated CD-epitopes.[139]
Methane sulfonateMutations in α- and γ-gliadin genesG/C to A/T nucleotides in the DNA transitionRemoving or reducing the toxic fractions of gluten.[127]
Plant
breeding
Less immunogenic alternative cropsTriticum monoccoumAncient wheat varieties that lack the D genome, known to encode highly immunogenic peptides.Includes significantly fewer toxic sequences.[120,140,141,142]
Synthetic hexaploid wheatHybridization of Triticum turgidum ssp. durum with Aegilops tauschii linesLow levels of CD-toxic gliadins.[125]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Di Stasio, L.; Mamone, G. Gluten Proteins: Beneficial Factors and Toxic Triggers in Human Health. Foods 2025, 14, 3403. https://doi.org/10.3390/foods14193403

AMA Style

Di Stasio L, Mamone G. Gluten Proteins: Beneficial Factors and Toxic Triggers in Human Health. Foods. 2025; 14(19):3403. https://doi.org/10.3390/foods14193403

Chicago/Turabian Style

Di Stasio, Luigia, and Gianfranco Mamone. 2025. "Gluten Proteins: Beneficial Factors and Toxic Triggers in Human Health" Foods 14, no. 19: 3403. https://doi.org/10.3390/foods14193403

APA Style

Di Stasio, L., & Mamone, G. (2025). Gluten Proteins: Beneficial Factors and Toxic Triggers in Human Health. Foods, 14(19), 3403. https://doi.org/10.3390/foods14193403

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop