1
|
Rahmani S, Galipeau HJ, Clarizio AV, Wang X, Hann A, Rueda GH, Kirtikar UN, Constante M, Wulczynski M, Su HM, Burchett R, Bramson JL, Pinto-Sanchez MI, Stefanolo JP, Niveloni S, Surette MG, Murray JA, Anderson RP, Bercik P, Caminero A, Chirdo FG, F Didar T, Verdu EF. Gluten-Dependent Activation of CD4 + T Cells by MHC Class II-Expressing Epithelium. Gastroenterology 2024; 167:1113-1128. [PMID: 39128638 DOI: 10.1053/j.gastro.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND & AIMS Intestinal epithelial cell (IEC) damage is a hallmark of celiac disease (CeD); however, its role in gluten-dependent T-cell activation is unknown. We investigated IEC-gluten-T-cell interactions in organoid monolayers expressing human major histocompatibility complex class II (HLA-DQ2.5), which facilitates gluten antigen recognition by CD4+ T cells in CeD. METHODS Epithelial major histocompatibility complex class II (MHCII) was determined in active and treated CeD, and in nonimmunized and gluten-immunized DR3-DQ2.5 transgenic mice, lacking mouse MHCII molecules. Organoid monolayers from DR3-DQ2.5 mice were treated with or without interferon (IFN)-γ, and MHCII expression was evaluated by flow cytometry. Organoid monolayers and CD4+ T-cell co-cultures were incubated with gluten, predigested, or not by elastase-producing Pseudomonas aeruginosa or its lasB mutant. T-cell function was assessed based on proliferation, expression of activation markers, and cytokine release in the co-culture supernatants. RESULTS Patients with active CeD and gluten-immunized DR3-DQ2.5 mice demonstrated epithelial MHCII expression. Organoid monolayers derived from gluten-immunized DR3-DQ2.5 mice expressed MHCII, which was upregulated by IFN-γ. In organoid monolayer T-cell co-cultures, gluten increased the proliferation of CD4+ T cells, expression of T-cell activation markers, and the release of interleukin-2, IFN-γ, and interleukin-15 in co-culture supernatants. Gluten metabolized by P aeruginosa, but not the lasB mutant, enhanced CD4+ T-cell proliferation and activation. CONCLUSIONS Gluten antigens are efficiently presented by MHCII-expressing IECs, resulting in the activation of gluten-specific CD4+ T cells, which is enhanced by gluten predigestion with microbial elastase. Therapeutics directed at IECs may offer a novel approach for modulating both adaptive and innate immunity in patients with CeD.
Collapse
Affiliation(s)
- Sara Rahmani
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alexandra V Clarizio
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Xuanyu Wang
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Amber Hann
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gaston H Rueda
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Utkarshini N Kirtikar
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Marco Constante
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mark Wulczynski
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hsuan-Ming Su
- Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Rebecca Burchett
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Maria Ines Pinto-Sanchez
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | - Michael G Surette
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Department of Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | | | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Fernando G Chirdo
- Instituto de Estudios Inmunológicos y Fisiopatológicos - IIFP (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.
| | - Tohid F Didar
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada; Department of Mechanical Engineering, McMaster University, Hamilton, Ontario, Canada.
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
2
|
Dayon L, Núñez Galindo A, Chevalier J, Aquarius M, Otten B, Troost FJ, Duncan P, Affolter M. Detection of gluten peptides in human duodenal fluids with immuno-liquid chromatography-mass spectrometry. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:6819-6828. [PMID: 39264106 DOI: 10.1039/d4ay00852a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Gluten proteins are storage proteins in wheat that exhibit a certain resistance to gastrointestinal digestion. To explore solutions to cope with accidental ingestion of gluten in individuals suffering from gluten-related disorders, it is essential to monitor the fate of gluten peptides in biological samples, i.e., gastrointestinal juices, blood plasma or urine. In this work, we aimed at developing a mass spectrometry (MS)-based method for measuring gluten peptides in human duodenal fluids. Seven gluten peptides, including the well-documented 33-mer gluten peptide (LQLQPFPQPQLPYPQPQLPYPQPQLPYPQPQPF), were selected after a literature review and characterization of a gluten-containing product. Isotopically labelled peptides were used as references and a targeted liquid chromatography (LC) MS assay based on high resolution parallel reaction monitoring (PRM) was designed. Despite iterative and fine tuning of the LC-PRM-MS method, the low level of endogenous gluten peptides in human duodenal fluid samples precluded their direct detection. Thus, an initial immunoprecipitation (IP) step was included. Several antibodies were tested, and one proved reliable for the enrichment of the 33-mer gluten peptide as well as a few additional gluten peptides. Figures-of-merits of the immuno-LC-PRM-MS assay were assessed with a focus on quantification trueness and precision. We have developed an MS-based method for measuring the 33-mer gluten peptide in human duodenal fluids. Based on isotopic dilution, the method relies on the combination of IP and LC-PRM-MS analysis. Measurements were shown to be sensitive, quantitative, and reproducible.
Collapse
Affiliation(s)
- Loïc Dayon
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Société des Produits Nestlé SA, EPFL Innovation Park, Bâtiment H, CH-1015 Lausanne, Switzerland.
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Antonio Núñez Galindo
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Société des Produits Nestlé SA, EPFL Innovation Park, Bâtiment H, CH-1015 Lausanne, Switzerland.
| | - Julien Chevalier
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Michèl Aquarius
- Department of Gastroenterology, VieCuri Medical Center, Venlo, The Netherlands
| | - Britt Otten
- Department of Human Biology, NUTRIM, Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation (HEFI), Maastricht University, Venlo, The Netherlands
| | - Freddy J Troost
- Department of Human Biology, NUTRIM, Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation (HEFI), Maastricht University, Venlo, The Netherlands
| | - Peter Duncan
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Michael Affolter
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Société des Produits Nestlé SA, EPFL Innovation Park, Bâtiment H, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
3
|
Rigo FF, Oliveira ECSD, Quaglio AEV, Moutinho BD, Di Stasi LC, Sassaki LY. Expression of MicroRNAs in Adults with Celiac Disease: A Narrative Review. Int J Mol Sci 2024; 25:9412. [PMID: 39273359 PMCID: PMC11395070 DOI: 10.3390/ijms25179412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/07/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy triggered by the ingestion of proline- and glutamine-rich proteins, widely termed "gluten", in genetically susceptible individuals. CD induces an altered immune response that leads to chronic inflammation and duodenal mucosal damage. Currently, there are no specific tests for the accurate diagnosis of CD, and no drugs are available to treat this condition. The only available treatment strategy is lifelong adherence to a gluten-free diet. However, some studies have investigated the involvement of microRNAs (miRNAs) in CD pathogenesis. miRNAs are small noncoding ribonucleic acid molecules that regulate gene expression. Despite the growing number of studies on the role of miRNAs in autoimmune disorders, data on miRNAs and CD are scarce. Therefore, this study aimed to perform a literature review to summarize CD, miRNAs, and the potential interactions between miRNAs and CD in adults. This review shows that miRNA expression can suppress or stimulate pathways related to CD pathogenesis by regulating cell proliferation and differentiation, regulatory T-cell development, innate immune response, activation of the inflammatory cascade, focal adhesion, T-cell commitment, tissue transglutaminase synthesis, and cell cycle. Thus, identifying miRNAs and their related effects on CD could open new possibilities for diagnosis, prognosis, and follow-up of biomarkers.
Collapse
Affiliation(s)
- Francielen Furieri Rigo
- Department of Internal Medicine, Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | | | | | - Bruna Damásio Moutinho
- Department of Gastroenterology, Division of Clinical Gastroenterology and Hepatology, University of São Paulo School of Medicine (USP), São Paulo 01246-903, SP, Brazil
| | - Luiz Claudio Di Stasi
- Laboratory of Phytomedicines, Pharmacology and Biotechnology (PhytoPharmaTec), Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - Ligia Yukie Sassaki
- Department of Internal Medicine, Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| |
Collapse
|
4
|
Liu YY, Ye RL, Meng M. Specificity Enhancement of Glutenase Bga1903 toward Celiac Disease-Eliciting Pro-Immunogenic Peptides via Active-Site Modification. Int J Mol Sci 2023; 25:505. [PMID: 38203677 PMCID: PMC10779176 DOI: 10.3390/ijms25010505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Celiac disease is an autoimmune disease triggered by oral ingestion of gluten, with certain gluten residues resistant to digestive tract enzymes. Within the duodenum, the remaining peptides incite immunogenic responses, including the generation of autoantibodies and inflammation, leading to irreversible damage. Our previous exploration unveiled a glutenase called Bga1903 derived from the Gram-negative bacterium Burkholderia gladioli. The cleavage pattern of Bga1903 indicates its moderate ability to mitigate the toxicity of pro-immunogenic peptides. The crystal structure of Bga1903, along with the identification of subsites within its active site, was determined. To improve its substrate specificity toward prevalent motifs like QPQ within gluten peptides, the active site of Bga1903 underwent site-directed mutagenesis according to structural insights and enzymatic kinetics. Among the double-site mutants, E380Q/S387L exhibits an approximately 34-fold increase in its specificity constant toward the QPQ sequence, favoring glutamines at the P1 and P3 positions compared to the wild type. The increased specificity of E380Q/S387L not only enhances its ability to break down pro-immunogenic peptides but also positions this enzyme variant as a promising candidate for oral therapy for celiac disease.
Collapse
Affiliation(s)
| | | | - Menghsiao Meng
- Graduate Institute of Biotechnology, National Chung Hsing University, 250 Kuo-Kuang Rd., Taichung 40227, Taiwan; (Y.-Y.L.); (R.-L.Y.)
| |
Collapse
|
5
|
Banerjee P, Chaudhary R, Singh AK, Parulekar P, Kumar S, Senapati S. Specific Genetic Polymorphisms Contributing in Differential Binding of Gliadin Peptides to HLA-DQ and TCR to Elicit Immunogenicity in Celiac Disease. Biochem Genet 2023; 61:2457-2480. [PMID: 37103600 DOI: 10.1007/s10528-023-10377-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 04/01/2023] [Indexed: 04/28/2023]
Abstract
Immunogenicity of gliadin peptides in celiac disease (CD) is majorly determined by the pattern of molecular interactions with HLA-DQ and T-cell receptors (TCR). Investigation of the interactions between immune-dominant gliadin peptides, DQ protein, and TCR are warranted to unravel the basis of immunogenicity and variability contributed by the genetic polymorphisms. Homology modeling of HLA and TCR done using Swiss Model and iTASSER, respectively. Molecular interactions of eight common deamidated immune-dominant gliadin with HLA-DQ allotypes and specific TCR gene pairs were evaluated. Docking of the three structures was performed with ClusPro2.0 and ProDiGY was used to predict binding energies. Effects of known allelic polymorphisms and reported susceptibility SNPs were predicted on protein-protein interactions. CD susceptible allele, HLA-DQ2.5 was shown to have considerable binding affinity to 33-mer gliadin (ΔG = - 13.9; Kd = 1.5E - 10) in the presence of TRAV26/TRBV7. Higher binding affinity was predicted (ΔG = - 14.3, Kd = 8.9E - 11) when TRBV28 was replaced with TRBV20 paired with TRAV4 suggesting its role in CD predisposition. SNP rs12722069 at HLA-DQ8 that codes Arg76α forms three H-bonds with Glu12 and two H-bonds with Asn13 of DQ2 restricted gliadin in the presence of TRAV8-3/TRBV6. None of the HLA-DQ polymorphisms was found to be in linkage disequilibrium with reported CD susceptibility markers. Haplotypic presentations of rs12722069-G, rs1130392-C, rs3188043-C and rs4193-A with CD reported SNPs were observed in sub-ethnic groups. Highly polymorphic sites of HLA alleles and TCR variable regions could be utilized for better risk prediction models in CD. Therapeutic strategies by identifying inhibitors or blockers targeting specific gliadin:HLA-DQ:TCR binding sites could be investigated.
Collapse
Affiliation(s)
- Pratibha Banerjee
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Ramprasad Chaudhary
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Atul Kumar Singh
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Pratima Parulekar
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Shashank Kumar
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, 151401, India.
| | - Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
6
|
Xiao B, Zhang C, Zhou J, Wang S, Meng H, Wu M, Zheng Y, Yu R. Design of SC PEP with enhanced stability against pepsin digestion and increased activity by machine learning and structural parameters modeling. Int J Biol Macromol 2023; 250:125933. [PMID: 37482154 DOI: 10.1016/j.ijbiomac.2023.125933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/20/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
Prolyl endopeptidases from Sphingomonas capsulata (SC PEP) has attracted much attention as promising oral therapy candidate for celiac sprue, however, its low stability in the gastric environment leads to unsatisfactory clinical results. Therefore, improving its stability against pepsin digestion at low pH is crucial for clinical applications, but challenging. In this study, machine learning and physical parameter model were combined to design SC PEP mutants. After iterations, 20 mutants had higher hydrolysis activity in stomach environment, which was up to 14.1-fold compared with wild-type SC PEP. Mutant M24 involving stable and active mutations and pegylated M24 (M24-PEG) had higher activity of hydrolyzing immunogen in bread than wild-type SC PEP in vitro and in vivo, and residual immunogens in simulated gastric environment were only 1/8 and 1/10 of that in the wild-type SC PEP group. The total residual immunogens in the gastrointestinal tract of mice in the M24 and M24-PEG groups were <20 ppm, reaching the standard of non-toxic food. Our results indicate that the combination of M24 (or M24-PEG) with EP-B2 may be a promising candidate for celiac disease, and the strategies developed in this study provide a paradigm for the design of SC PEP stability mutants.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Chun Zhang
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Junxiu Zhou
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Sa Wang
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Huan Meng
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Miao Wu
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China
| | - Yongxiang Zheng
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China.
| | - Rong Yu
- Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
7
|
Marín-Sanz M, Barro F, Sánchez-León S. Unraveling the celiac disease-related immunogenic complexes in a set of wheat and tritordeum genotypes: implications for low-gluten precision breeding in cereal crops. FRONTIERS IN PLANT SCIENCE 2023; 14:1171882. [PMID: 37251754 PMCID: PMC10210591 DOI: 10.3389/fpls.2023.1171882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/06/2023] [Indexed: 05/31/2023]
Abstract
The development of low-gluten immunogenic cereal varieties is a suitable way to fight the increment of pathologies associated with the consumption of cereals. Although RNAi and CRISPR/Cas technologies were effective in providing low-gluten wheat, the regulatory framework, particularly in the European Union, is an obstacle to the short- or medium-term implementation of such lines. In the present work, we carried out a high throughput amplicon sequencing of two highly immunogenic complexes of wheat gliadins in a set of bread and durum wheat, and tritordeum genotypes. Bread wheat genotypes harboring the 1BL/1RS translocation were included in the analysis and their amplicons successfully identified. The number of CD epitopes and their abundances were determined in the alpha- and gamma-gliadin amplicons, including 40k-γ-secalin ones. Bread wheat genotypes not containing the 1BL/1RS translocation showed a higher average number of both alpha- and gamma-gliadin epitopes than those containing such translocation. Interestingly, alpha-gliadin amplicons not containing CD epitopes accounted for the highest abundance (around 53%), and the alpha- and gamma-gliadin amplicons with the highest number of epitopes were present in the D-subgenome. The durum wheat and tritordeum genotypes showed the lowest number of alpha- and gamma-gliadin CD epitopes. Our results allow progress in unraveling the immunogenic complexes of alpha- and gamma-gliadins and can contribute to the development of low-immunogenic varieties within precision breeding programs, by crossing or by CRISPR/Cas gene editing.
Collapse
|
8
|
Auricchio R, Galatola M, Cielo D, Rotondo R, Carbone F, Mandile R, Carpinelli M, Vitale S, Matarese G, Gianfrani C, Troncone R, Auricchio S, Greco L. Antibody Profile, Gene Expression and Serum Cytokines in At-Risk Infants before the Onset of Celiac Disease. Int J Mol Sci 2023; 24:ijms24076836. [PMID: 37047806 PMCID: PMC10095049 DOI: 10.3390/ijms24076836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 03/23/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023] Open
Abstract
Immunological events that precede the development of villous atrophy in celiac disease (CeD) are still not completely understood. We aimed to explore CeD-associated antibody production (anti-native gliadin (AGA), anti-deamidated gliadin (DGP) and anti-tissue transglutaminase (anti-tTG)) in infants at genetic risk for CeD from the Italian cohorts of the PREVENT-CD and Neocel projects, as well as the relationship between antibody production and systemic inflammation. HLA DQ2 and/or DQ8 infants from families with a CeD case were followed from birth. Out of 220 at-risk children, 182 had not developed CeD by 6 years of age (CTRLs), and 38 developed celiac disease (CeD). The profiles of serum cytokines (INFγ, IL1β, IL2, IL4, IL6, IL10, IL12p70, IL17A and TNFα) and the expression of selected genes (FoxP3, IL10, TGFβ, INFγ, IL4 and IL2) were evaluated in 46 children (20 CeD and 26 CTRLs). Among the 182 healthy CTRLs, 28 (15.3%) produced high levels of AGA-IgA (AGA+CTRLs), and none developed anti-tTG-IgA or DGP-IgA, compared to 2/38 (5.3%) CeD infants (Chi Sq. 5.97, p = 0.0014). AGAs appeared earlier in CTRLs than in those who developed CeD (19 vs. 28 months). Additionally, the production of AGAs in CeD overlapped with the production of DGP and anti-tTG. In addition, gene expression as well as serum cytokine levels discriminated children who developed CeD from CTRLs. In conclusion, these findings suggest that the early and isolated production of AGA-IgA antibodies is a CeD-tolerogenic marker and that changes in gene expression and cytokine patterns precede the appearance of anti-tTG antibodies.
Collapse
Affiliation(s)
- Renata Auricchio
- Department of Translational Medical Science, University Federico II, 80131 Naples, Italy
- European Laboratory for Food Induced Diseases, University Federico II, 80131 Naples, Italy
| | - Martina Galatola
- Department of Translational Medical Science, University Federico II, 80131 Naples, Italy
| | - Donatella Cielo
- Department of Translational Medical Science, University Federico II, 80131 Naples, Italy
| | - Roberta Rotondo
- Department of Translational Medical Science, University Federico II, 80131 Naples, Italy
| | - Fortunata Carbone
- Laboratory of Immunology, Institute for Experimental Endocrinology and Oncology, National Research Council of Italy (IEOS-CNR), c/o Department of Molecular Medicine and Medical Biotechnology, University Federico II, 80131 Naples, Italy
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Roberta Mandile
- Department of Translational Medical Science, University Federico II, 80131 Naples, Italy
- European Laboratory for Food Induced Diseases, University Federico II, 80131 Naples, Italy
| | - Martina Carpinelli
- Department of Translational Medical Science, University Federico II, 80131 Naples, Italy
- European Laboratory for Food Induced Diseases, University Federico II, 80131 Naples, Italy
| | - Serena Vitale
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), 80131 Naples, Italy
| | - Giuseppe Matarese
- Laboratory of Immunology, Institute for Experimental Endocrinology and Oncology, National Research Council of Italy (IEOS-CNR), c/o Department of Molecular Medicine and Medical Biotechnology, University Federico II, 80131 Naples, Italy
| | - Carmen Gianfrani
- European Laboratory for Food Induced Diseases, University Federico II, 80131 Naples, Italy
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), 80131 Naples, Italy
| | - Riccardo Troncone
- Department of Translational Medical Science, University Federico II, 80131 Naples, Italy
- European Laboratory for Food Induced Diseases, University Federico II, 80131 Naples, Italy
| | - Salvatore Auricchio
- European Laboratory for Food Induced Diseases, University Federico II, 80131 Naples, Italy
| | - Luigi Greco
- European Laboratory for Food Induced Diseases, University Federico II, 80131 Naples, Italy
| |
Collapse
|
9
|
Impacts of Sourdough Technology on the Availability of Celiac Peptides from Wheat α- and γ-Gliadins: In Silico Approach. ALLERGIES 2023. [DOI: 10.3390/allergies3010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Celiac peptide-generating α- and γ-gliadins consist of a disordered N-terminal domain extended by an α-helical-folded C-terminal domain. Celiac peptides, primarily located along the disordered part of α- and γ-gliadin molecules, are nicely exposed and directly accessible to proteolytic enzymes occurring in the gastric (pepsin) and intestinal (trypsin, chymotrypsin) fluids. More than half of the potential celiac peptides identified so far in gliadins exhibit cleavage sites for pepsin. However, celiac peptides proteolytically truncated by one or two amino acid residues could apparently retain some activity toward HLA-DQ2 and HLA-DQ8 receptors in docking experiments. Together with the uncleaved peptides, these still active partially degraded CD peptides account for the incapacity of the digestion process to inactivate CD peptides from gluten proteins. In contrast, sourdough fermentation processes involve other proteolytic enzymes susceptible to the deep degradation of celiac peptides. In particular, sourdough supplemented by fungal prolyl endoproteases enhances the degrading capacities of the sourdough fermentation process toward celiac peptides. Nevertheless, since tiny amounts of celiac peptides sufficient to trigger deleterious effects on CD people can persist in sourdough-treated bread and food products, it is advisable to avoid consumption of sourdough-treated food products for people suffering from celiac disease. As an alternative, applying the supplemented sourdough process to genetically modified low gluten or celiac-safe wheat lines should result in food products that are safer for susceptible and CD people.
Collapse
|
10
|
Human glycoprotein-2 expressed in Brunner glands - A putative autoimmune target and link between Crohn's and coeliac disease. Clin Immunol 2023; 247:109214. [PMID: 36608744 DOI: 10.1016/j.clim.2022.109214] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023]
Abstract
Glycoprotein 2 (GP2) is an autoantigen in Crohn's (CD) and coeliac disease (CeD). We assessed GP2-isoform (GP21-4)-expression in intestinal biopsies of paediatric patients with CD, CeD, ulcerative colitis (UC), and healthy children (HC). Transcription of GP21-4 was elevated in proximal small intestine in CeD and CD patients (only GP22/4) compared to jejunum (CeD/CD) and large bowel (CD). CeD patients demonstrated higher duodenal GP22/4-mRNA levels compared to HC/UC patients whereas CD patients showed higher GP24-mRNA levels compared to UC patients. Duodenal synthesis of only small GP2 isoforms (GP23/4) was demonstrated in epithelial cells in patients/HC and in Brunner glands (also large isoforms) with a more frequent apical location in CD/CeD patients. All four GP2 isoforms interacted with gliadin and phosphopeptidomannan. Gliadin digestion improved binding to GP2 isoforms. GP21-4 binding to CeD/CD-related antigens, elevated duodenal GP21-4-mRNA transcription, and GP2-protein secretion in Brunner glands of CeD/CD patients suggest an autoimmune CeD/CD link.
Collapse
|
11
|
Probiotics in the Sourdough Bread Fermentation: Current Status. FERMENTATION 2023. [DOI: 10.3390/fermentation9020090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Sourdough fermentation is an ancient technique to ferment cereal flour that improves bread quality, bringing nutritional and health benefits. The fermented dough has a complex microbiome composed mainly of lactic acid bacteria and yeasts. During fermentation, the production of metabolites and chemical reactions occur, giving the product unique characteristics and a high sensory quality. Mastery of fermentation allows adjustment of gluten levels, delaying starch digestibility, and increasing the bio-accessibility of vitamins and minerals. This review focuses on the main steps of sourdough fermentation, the microorganisms involved, and advances in bread production with functional properties. The impact of probiotics on human health, the metabolites produced, and the main microbial enzymes used in the bakery industry are also discussed.
Collapse
|
12
|
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. [PMID: 36741992 PMCID: PMC9890883 DOI: 10.3389/fnut.2022.1049623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/15/2022] [Indexed: 01/19/2023] Open
Abstract
Gluten proteins are the causative agents of celiac disease (CD), a lifelong and worldwide spread food intolerance, characterized by an autoimmune enteropathy. Gluten is a complex mixture of high homologous water-insoluble proteins, characterized by a high content of glutamine and proline amino acids that confers a marked resistance to degradation by gastrointestinal proteases. As a consequence of that, large peptides are released in the gut lumen with the potential to activate inflammatory T cells, in CD predisposed individuals. To date, several strategies aimed to detoxify gluten proteins or to develop immunomodulatory drugs to recover immune tolerance to gluten are under investigation. This review overviews the state of art of both analytical and functional methods currently used to assess the immunogenicity potential of gluten proteins from different cereal sources, including native raw seed flours and complex food products, as well as drug-treated samples. The analytical design to assess the content and profile of gluten immunogenic peptides, described herein, is based on the oral-gastro-intestinal digestion (INFOGEST model) followed by extensive characterization of residual gluten peptides by proteomic and immunochemical analyses. These approaches include liquid chromatography-high-resolution mass spectrometry (LC-MS/MS) and R5/G12 competitive ELISA. Functional studies to assess the immune stimulatory capabilities of digested gluten peptides are based on gut mucosa T cells or peripheral blood cells obtained from CD volunteers after a short oral gluten challenge.
Collapse
Affiliation(s)
- Gianfranco Mamone
- Institute of Food Science, Department of Biology, Agriculture and Food Sciences, National Research Council of Italy, Avellino, Italy
| | - Luigia Di Stasio
- Institute of Food Science, Department of Biology, Agriculture and Food Sciences, National Research Council of Italy, Avellino, Italy
| | - Serena Vitale
- Institute of Biochemistry and Cell Biology, Department of Biomedical Sciences, National Research Council of Italy, Naples, Italy
| | - Stefania Picascia
- Institute of Biochemistry and Cell Biology, Department of Biomedical Sciences, National Research Council of Italy, Naples, Italy
| | - Carmen Gianfrani
- Institute of Biochemistry and Cell Biology, Department of Biomedical Sciences, National Research Council of Italy, Naples, Italy,*Correspondence: Carmen Gianfrani,
| |
Collapse
|
13
|
Vinzant K, Rashid M, Khodakovskaya MV. Advanced applications of sustainable and biological nano-polymers in agricultural production. FRONTIERS IN PLANT SCIENCE 2023; 13:1081165. [PMID: 36684740 PMCID: PMC9852866 DOI: 10.3389/fpls.2022.1081165] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/08/2022] [Indexed: 06/17/2023]
Abstract
Though still in its infancy, the use of nanotechnology has shown promise for improving and enhancing agriculture: nanoparticles (NP) offer the potential solution to depleted and dry soils, a method for the controlled release of agrochemicals, and offer an easier means of gene editing in plants. Due to the continued growth of the global population, it is undeniable that our agricultural systems and practices will need to become more efficient in the very near future. However, this new technology comes with significant worry regarding environmental contamination. NP applied to soils could wash into aquifers and contaminate drinking water, or NP applied to food crops may carry into the end product and contaminate our food supply. These are valid concerns that are not likely to be fully answered in the immediate future due to the complexity of soil-NP interactions and other confounding variables. Therefore, it is obviously preferred that NP used outdoors at this early stage be biodegradable, non-toxic, cost-effective, and sustainably manufactured. Fortunately, there are many different biologically derived, cost-efficient, and biocompatible polymers that are suitable for agricultural applications. In this mini-review, we discuss some promising organic nanomaterials and their potential use for the optimization and enhancement of agricultural practices.
Collapse
|
14
|
Complex of Proline-Specific Peptidases in the Genome and Gut Transcriptomes of Tenebrionidae Insects and Their Role in Gliadin Hydrolysis. Int J Mol Sci 2022; 24:ijms24010579. [PMID: 36614021 PMCID: PMC9820350 DOI: 10.3390/ijms24010579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022] Open
Abstract
A detailed analysis of the complexes of proline-specific peptidases (PSPs) in the midgut transcriptomes of the larvae of agricultural pests Tenebrio molitor and Tribolium castaneum and in the genome of T. castaneum is presented. Analysis of the T. castaneum genome revealed 13 PSP sequences from the clans of serine and metal-dependent peptidases, of which 11 sequences were also found in the gut transcriptomes of both tenebrionid species' larvae. Studies of the localization of PSPs, evaluation of the expression level of their genes in gut transcriptomes, and prediction of the presence of signal peptides determining secretory pathways made it possible to propose a set of peptidases that can directly participate in the hydrolysis of food proteins in the larvae guts. The discovered digestive PSPs of tenebrionids in combination with the post-glutamine cleaving cysteine cathepsins of these insects effectively hydrolyzed gliadins, which are the natural food substrates of the studied pests. Based on the data obtained, a hypothetical scheme for the complete hydrolysis of immunogenic gliadin peptides by T. molitor and T. castaneum digestive peptidases was proposed. These results show promise regarding the development of a drug based on tenebrionid digestive enzymes for the enzymatic therapy of celiac disease and gluten intolerance.
Collapse
|
15
|
Wagh SK, Lammers KM, Padul MV, Rodriguez-Herrera A, Dodero VI. Celiac Disease and Possible Dietary Interventions: From Enzymes and Probiotics to Postbiotics and Viruses. Int J Mol Sci 2022; 23:ijms231911748. [PMID: 36233048 PMCID: PMC9569549 DOI: 10.3390/ijms231911748] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/24/2022] Open
Abstract
Celiac Disease (CeD) is a chronic small intestinal immune-mediated enteropathy caused by the ingestion of dietary gluten proteins in genetically susceptible individuals. CeD is one of the most common autoimmune diseases, affecting around 1.4% of the population globally. To date, the only acceptable treatment for CeD is strict, lifelong adherence to a gluten-free diet (GFD). However, in some cases, GFD does not alter gluten-induced symptoms. In addition, strict adherence to a GFD reduces patients’ quality of life and is often a socio-economic burden. This narrative review offers an interdisciplinary overview of CeD pathomechanism and the limitations of GFD, focusing on current research on possible dietary interventions. It concentrates on the recent research on the degradation of gluten through enzymes, the modulation of the microbiome, and the different types of “biotics” strategies, from probiotics to the less explored “viromebiotics” as possible beneficial complementary interventions for CeD management. The final aim is to set the context for future research that may consider the role of gluten proteins and the microbiome in nutritional and non-pharmacological interventions for CeD beyond the sole use of the GFD.
Collapse
Affiliation(s)
- Sandip K. Wagh
- Department of Organic and Bioorganic Chemistry, Bielefeld University, 33615 Bielefeld, Germany
- Department of Biochemistry, Dr. Babasaheb Ambedkar Marathwada University, Aurangabad 431004, India
| | | | - Manohar V. Padul
- Department of Biochemistry, The Institute of Science, Dr. Homi Bhabha State University, Mumbai 400032, India
| | | | - Veronica I. Dodero
- Department of Organic and Bioorganic Chemistry, Bielefeld University, 33615 Bielefeld, Germany
- Correspondence:
| |
Collapse
|
16
|
Wagh SK, Lammers KM, Padul MV, Rodriguez-Herrera A, Dodero VI. Celiac Disease and Possible Dietary Interventions: From Enzymes and Probiotics to Postbiotics and Viruses. Int J Mol Sci 2022. [PMID: 36233048 DOI: 10.3390/ijms231911748.pmid:36233048;pmcid:pmc9569549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
Celiac Disease (CeD) is a chronic small intestinal immune-mediated enteropathy caused by the ingestion of dietary gluten proteins in genetically susceptible individuals. CeD is one of the most common autoimmune diseases, affecting around 1.4% of the population globally. To date, the only acceptable treatment for CeD is strict, lifelong adherence to a gluten-free diet (GFD). However, in some cases, GFD does not alter gluten-induced symptoms. In addition, strict adherence to a GFD reduces patients' quality of life and is often a socio-economic burden. This narrative review offers an interdisciplinary overview of CeD pathomechanism and the limitations of GFD, focusing on current research on possible dietary interventions. It concentrates on the recent research on the degradation of gluten through enzymes, the modulation of the microbiome, and the different types of "biotics" strategies, from probiotics to the less explored "viromebiotics" as possible beneficial complementary interventions for CeD management. The final aim is to set the context for future research that may consider the role of gluten proteins and the microbiome in nutritional and non-pharmacological interventions for CeD beyond the sole use of the GFD.
Collapse
Affiliation(s)
- Sandip K Wagh
- Department of Organic and Bioorganic Chemistry, Bielefeld University, 33615 Bielefeld, Germany
- Department of Biochemistry, Dr. Babasaheb Ambedkar Marathwada University, Aurangabad 431004, India
| | | | - Manohar V Padul
- Department of Biochemistry, The Institute of Science, Dr. Homi Bhabha State University, Mumbai 400032, India
| | | | - Veronica I Dodero
- Department of Organic and Bioorganic Chemistry, Bielefeld University, 33615 Bielefeld, Germany
| |
Collapse
|
17
|
Liu YY, Lin IC, Chen PC, Lee CC, Meng M. Crystal structure of a Burkholderia peptidase and modification of the substrate-binding site for enhanced hydrolytic activity toward gluten-derived pro-immunogenic peptides. Int J Biol Macromol 2022; 222:2258-2269. [DOI: 10.1016/j.ijbiomac.2022.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/19/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
|
18
|
Mamone G, Comelli MC, 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. [PMID: 36159465 PMCID: PMC9493446 DOI: 10.3389/fnut.2022.974771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
Gluten degrading enzymes, which are commonly referred to as “glutenases,” represent attractive candidates for the development of a pharmacological treatment of gluten related disorders, such as coeliac disease (CeD). Endoprotease-40 (E40), a novel glutenase secreted by the actinomycete Actinoallomurus A8 and recombinantly produced in S. lividans TK24, was shown to be active at pH 3 to 6 (optimum pH 5), resistant to pepsin and trypsin degradation, able to destroy immunotoxicity of both gliadin 33-mer peptide and whole proteins and to strongly reduce the response of specific T cells when added to gliadin in in vitro gastrointestinal digestion. This study aims to functionally assess the capabilities of Endoprotease-40 (E40) to detoxify residual gluten immunogenic peptides in gastrointestinal digesta of food matrices made of soft and durum wheat. The INFOGEST harmonized protocols were applied to the multicompartmental model of simulated human gastrointestinal digestion, for the quantitative assessment of residual gluten in liquid (beer) and solid (bread and pasta) foods, made of either soft or durum wheat. Proteomic and immunological techniques, and functional assays on intestinal T cell lines from celiac disease patients were used to identify gluten-derived immunogenic peptide sequences surviving in gastric and gastrointestinal digesta after the addition of E40 at increasing enzyme: wheat proteins ratios. During the gastric phase (2 h incubation time), the addition of E40 demonstrated an extensive (≥ 95%) dose-dependent detoxification of whole gluten in real food matrices. Overall, the residual gluten content was found at, or even below, the 20 ppm gluten-free threshold for soft and durum wheat-based food. Furthermore, unlike in untreated gastrointestinal digesta, none of the immunodominant α-gliadin peptides survived in E40-treated digesta. Traces of ω- and γ-gliadin derived immunogenic peptides were still detected in E40-treated digesta, but unable to stimulate celiac-intestinal T cells. In conclusion, E40 is a promising candidate for the oral enzymatic therapy of CeD, as a stand-alone enzyme being efficient along the complete gastrointestinal digestion of gluten.
Collapse
Affiliation(s)
- Gianfranco Mamone
- Institute of Food Science, National Research Council of Italy, Avellino, Italy
- *Correspondence: Gianfranco Mamone,
| | | | - Serena Vitale
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| | - Luigia Di Stasio
- Institute of Food Science, National Research Council of Italy, Avellino, Italy
| | | | - Ilaria Mottola
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| | - Francesco Siano
- Institute of Food Science, National Research Council of Italy, Avellino, Italy
| | - Linda Cavaletti
- Fondazione Istituto Insubrico Ricerca per la Vita, Varese, Italy
| | - Carmen Gianfrani
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| |
Collapse
|
19
|
Qasim H, Nasr M, Mohammad A, Hor M, Baradeiya AM. Dysbiosis and Migraine Headaches in Adults With Celiac Disease. Cureus 2022; 14:e28346. [PMID: 36168375 PMCID: PMC9506300 DOI: 10.7759/cureus.28346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
One of the most significant illnesses associated with gluten is celiac disease, which encompasses many conditions. It is generally recognized that neurological manifestations can occur either at the time of the disease onset or as the illness continues to develop. One of the main clinical presentations of celiac disease is headache, either in the form of migraine or in an unspecific form. Migraine pathophysiology is intricate and still poorly understood. Several mechanisms involving the gut-brain axis have been proposed to explain this association. These include the interaction of chronic inflammation with inflammatory and vasoactive mediators, the modulation of the intestinal immune environment of the microbiota, and the dysfunction of the autonomic nervous system. However, further research is required to fully comprehend the fundamental mechanisms and pathways at play. This review aims to give a narrative summary of the literature on celiac disease's neurological symptoms, particularly migraines, and to assess any potential associations to dysbiosis, an imbalance in the microbiome.
Collapse
|
20
|
Recombinant Cathepsin L of Tribolium castaneum and Its Potential in the Hydrolysis of Immunogenic Gliadin Peptides. Int J Mol Sci 2022; 23:ijms23137001. [PMID: 35806001 PMCID: PMC9266932 DOI: 10.3390/ijms23137001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 02/04/2023] Open
Abstract
Wheat gliadins contain a large amount of glutamine- and proline-rich peptides which are not hydrolyzed by human digestive peptidases and can cause autoimmune celiac disease and other forms of gluten intolerance in predisposed people. Peptidases that efficiently cleave such immunogenic peptides can be used in enzyme therapy. The stored product insect pest Tribolium castaneum efficiently hydrolyzes gliadins. The main digestive peptidase of T. castaneum is cathepsin L, which is from the papain C1 family with post-glutamine cleavage activity. We describe the isolation and characterization of T. castaneum recombinant procathepsin L (rpTcCathL1, NP_001164001), which was expressed in Pichia pastoris cells. The activation of the proenzyme was conducted by autocatalytic processing. The effects of pH and proenzyme concentration in the reaction mixture on the processing were studied. The mature enzyme retained high activity in the pH range from 5.0 to 9.0 and displayed high pH-stability from 4.0 to 8.0 at 20 °C. The enzyme was characterized according to electrophoretic mobility under native conditions, activity and stability at various pH values, a sensitivity to various inhibitors, and substrate specificity, and its hydrolytic effect on 8-, 10-, 26-, and 33-mer immunogenic gliadins peptides was demonstrated. Our results show that rTcCathL1 is an effective peptidase that can be used to develop a drug for the enzyme therapy of various types of gluten intolerance.
Collapse
|
21
|
Functional implications of the CpG island methylation in the pathogenesis of celiac disease. Mol Biol Rep 2022; 49:10051-10064. [PMID: 35633417 DOI: 10.1007/s11033-022-07585-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/09/2022] [Indexed: 10/18/2022]
Abstract
Investigation of gene-environment cross talk through epigenetic modifications led to better understanding of the number of complex diseases. Clinical heterogeneity and differential treatment response often contributed by the epigenetic signatures which could be personal. DNA methylation at CpG islands presents a critical nuclear process as a result of gene-environment interactions. These CpG islands are frequently present near the promoter sequence of genes and get differentially methylated under specific environmental conditions. Technical advancements facilitate in high throughput screening of differentially methylated CpG islands. Recent epigenetic studies unraveled several CD susceptibility genes expressed in peripheral blood lymphocytes (PBLs), duodenal mucosa, lamina and epithelial cells that are influenced by differentially methylated CpG islands. Here we highlighted these susceptibility genes; classify these genes based on cellular functions and tissue of expression. We further discussed how these genes interacts with each other to influence critical pathways like NF-κB signaling pathway, IL-17 signaling cascade, RIG-I like receptor signaling pathway, NOD-like receptor pathways among several others. This review also shed light on how gut microbiota may lead to the differential methylation of CpG islands of CD susceptibility genes. Large scale epigenetic studies followed by estimation of heritability of these CpG methylation and polygenic risk score estimation of these genes would prioritize potentially druggable targets for better therapeutics. In vivo studies are warranted to unravel further cellular responses to CpG methylation.
Collapse
|
22
|
Büchold C, Hils M, Gerlach U, Weber J, Pelzer C, Heil A, Aeschlimann D, Pasternack R. Features of ZED1227: The First-In-Class Tissue Transglutaminase Inhibitor Undergoing Clinical Evaluation for the Treatment of Celiac Disease. Cells 2022; 11:cells11101667. [PMID: 35626704 PMCID: PMC9139979 DOI: 10.3390/cells11101667] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
ZED1227 is a small molecule tissue transglutaminase (TG2) inhibitor. The compound selectively binds to the active state of TG2, forming a stable covalent bond with the cysteine in its catalytic center. The molecule was designed for the treatment of celiac disease. Celiac disease is an autoimmune-mediated chronic inflammatory condition of the small intestine affecting about 1–2% of people in Caucasian populations. The autoimmune disease is triggered by dietary gluten. Consumption of staple foods containing wheat, barley, or rye leads to destruction of the small intestinal mucosa in genetically susceptible individuals, and this is accompanied by the generation of characteristic TG2 autoantibodies. TG2 plays a causative role in the pathogenesis of celiac disease. Upon activation by Ca2+, it catalyzes the deamidation of gliadin peptides as well as the crosslinking of gliadin peptides to TG2 itself. These modified biological structures trigger breaking of oral tolerance to gluten, self-tolerance to TG2, and the activation of cytotoxic immune cells in the gut mucosa. Recently, in an exploratory proof-of-concept study, ZED1227 administration clinically validated TG2 as a “druggable” target in celiac disease. Here, we describe the specific features and profiling data of the drug candidate ZED1227. Further, we give an outlook on TG2 inhibition as a therapeutic approach in indications beyond celiac disease.
Collapse
Affiliation(s)
- Christian Büchold
- Zedira GmbH, Roesslerstrasse 83, 64293 Darmstadt, Germany; (C.B.); (M.H.); (J.W.); (C.P.); (A.H.)
| | - Martin Hils
- Zedira GmbH, Roesslerstrasse 83, 64293 Darmstadt, Germany; (C.B.); (M.H.); (J.W.); (C.P.); (A.H.)
| | - Uwe Gerlach
- Sanofi-Aventis Deutschland GmbH, UG Serves as External Consultant for Medicinal Chemistry to Zedira, 65926 Frankfurt, Germany;
| | - Johannes Weber
- Zedira GmbH, Roesslerstrasse 83, 64293 Darmstadt, Germany; (C.B.); (M.H.); (J.W.); (C.P.); (A.H.)
| | - Christiane Pelzer
- Zedira GmbH, Roesslerstrasse 83, 64293 Darmstadt, Germany; (C.B.); (M.H.); (J.W.); (C.P.); (A.H.)
| | - Andreas Heil
- Zedira GmbH, Roesslerstrasse 83, 64293 Darmstadt, Germany; (C.B.); (M.H.); (J.W.); (C.P.); (A.H.)
| | - Daniel Aeschlimann
- Matrix Biology & Tissue Repair Research Unit, School of Dentistry, Cardiff University, Heath Park, Cardiff CF14 4XY, Wales, UK;
| | - Ralf Pasternack
- Zedira GmbH, Roesslerstrasse 83, 64293 Darmstadt, Germany; (C.B.); (M.H.); (J.W.); (C.P.); (A.H.)
- Correspondence:
| |
Collapse
|
23
|
Novel Gluten-Free Cinnamon Rolls by Substituting Wheat Flour with Resistant Starch, Lupine and Flaxseed Flour. Foods 2022; 11:foods11071022. [PMID: 35407109 PMCID: PMC8998033 DOI: 10.3390/foods11071022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Celiac disease (CD) is an immunological mediated disorder that occurs to genetically susceptible individuals who suffer from gluten consumption. Therefore, the most effective treatment of CD is a life-long gluten-free diet. This study aimed to produce a nutritious gluten-free cinnamon roll, where resistant starch and lupine flour were used instead of wheat flour, in addition to 10% flaxseed flour and a fixed amount of hydrocolloid (1% xanthan gum). Eight different gluten-free cinnamon roll treatments (T1–T8) were produced with different ratios of resistant starch and lupine flour according to the following percentages (85:5, 80:10, 75:15, 70:20, 65:25, 60:30, 55:35 and 50:40, respectively). The proximate analysis, physical properties, color measurements and sensory evaluation of all cinnamon roll treatments and flours were determined. It was found that lupine and flaxseed flours in all different treatments had significantly (p ≤ 0.05) higher levels of ash, protein, lipid and crude fiber compared to wheat flour treatment (control treatment). However, carbohydrate levels were significantly (p ≤ 0.05) higher in control treatment compared with treatments 3–8. Gluten-free cinnamon rolls had significantly (p ≤ 0.05) higher levels of unsaturated fatty acids (oleic acid, linoleic acid and linolenic acid) than control. Further, there were significant differences in lightness (L*), redness (a*) and yellowness (b*) color values between the gluten-free and control treatments. The control cinnamon roll significantly (p ≤ 0.05) had the highest level of lightness and the lowest level of redness. The sensory evaluation obtained by consumer evaluation indicated that control cinnamon rolls significantly (p ≤ 0.05) received the highest score in overall impression, overall flavor, hardness and aftertaste. However, treatment 5 significantly (p ≤ 0.05) received the highest score in all the sensory scores in comparison with other gluten-free treatments. It is possible to develop a quality gluten-free cinnamon roll with respect to nutritional value manifested in higher levels of protein, fibers, unsaturated acids and prebiotics with acceptable sensory attributes.
Collapse
|
24
|
Changes in Non-Deamidated versus Deamidated Epitope Targeting and Disease Prediction during the Antibody Response to Gliadin and Transglutaminase of Infants at Risk for Celiac Disease. Int J Mol Sci 2022; 23:ijms23052498. [PMID: 35269639 PMCID: PMC8909931 DOI: 10.3390/ijms23052498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Celiac disease (CeD) is a conditional autoimmune disorder with T cell-mediated immune response to gluten coupled with antibody production to gliadin and the self-protein tissue transglutaminase (TG2). TG2 contributes to the CeD pathomechanism by deamidating gliadin, thereby generating more immunogenic peptides. Anti-gliadin antibodies may appear before the autoantibody production. The scope of this study was to dissect these early antibody responses by investigating serum samples collected during the PreventCD prospective double-blind study, where infants with high CeD risk were randomized to 200 mg daily gluten intake or placebo from 4 to 6 months of age, followed by frequent blood testing on regular gluten consumption in both groups. After primary gluten intake, children with or without later CeD produced IgA and IgG antibodies which preferentially recognized non-deamidated gliadin peptides. At CeD development with anti-TG2 seroconversion, there was a significant increase in the antibody reaction toward deamidated gliadin peptides (DGP), with maturation in the binding strength for the PEQPFP gamma-gliadin core peptide. The earliest produced autoantibodies targeted TG2’s celiac epitope 2. Our results reveal a qualitative change in the gliadin-directed humoral immune response at the time when anti-TG2 antibodies appear, but anti-DGP antibodies in the absence of anti-TG2 antibodies are not disease-predictive.
Collapse
|
25
|
An efficient urine peptidomics workflow identifies chemically defined dietary gluten peptides from patients with celiac disease. Nat Commun 2022; 13:888. [PMID: 35173144 PMCID: PMC8850430 DOI: 10.1038/s41467-022-28353-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 01/20/2022] [Indexed: 12/11/2022] Open
Abstract
Celiac disease (CeD) is an autoimmune disorder induced by consuming gluten proteins from wheat, barley, and rye. Glutens resist gastrointestinal proteolysis, resulting in peptides that elicit inflammation in patients with CeD. Despite well-established connections between glutens and CeD, chemically defined, bioavailable peptides produced from dietary proteins have never been identified from humans in an unbiased manner. This is largely attributable to technical challenges, impeding our knowledge of potentially diverse peptide species that encounter the immune system. Here, we develop a liquid chromatographic-mass spectrometric workflow for untargeted sequence analysis of the urinary peptidome. We detect over 600 distinct dietary peptides, of which ~35% have a CeD-relevant T cell epitope and ~5% are known to stimulate innate immune responses. Remarkably, gluten peptides from patients with CeD qualitatively and quantitatively differ from controls. Our results provide a new foundation for understanding gluten immunogenicity, improving CeD management, and characterizing the dietary and urinary peptidomes. Gluten peptides from wheat enter the bloodstream and are excreted in urine but are yet to be chemically characterised. Here, the authors show by mass spectrometry that quantitative and qualitative differences in urinary peptides can be detected between healthy people and patients with celiac disease.
Collapse
|
26
|
Gliadin proteolytical resistant peptides: the interplay between structure and self-assembly in gluten-related disorders. Biophys Rev 2022; 13:1147-1154. [PMID: 35047092 PMCID: PMC8724473 DOI: 10.1007/s12551-021-00856-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/14/2021] [Indexed: 11/29/2022] Open
Abstract
In recent years, the evaluation of the structural properties of food has become of crucial importance in the understanding of food-related disorders. One of the most exciting systems is gliadin, a protein in wheat gluten, that plays a protagonist role in gluten-related disorders with a worldwide prevalence of 5%, including autoimmune celiac disease (CeD) (1%) and non-celiac wheat sensitivity (0.5–13%). It is accepted that gliadin is not fully digested by humans, producing large peptides that reach the gut mucosa. The gliadin peptides cross the lamina propria eliciting different immune responses in susceptible patients. Many clinical and biomedical efforts aim to diagnose and understand gluten-related disorders; meanwhile, the early stages of the inflammatory events remain elusive. Interestingly, although the primary sequence of many gliadin peptides is well known, it was only recently revealed the self-assembly capability of two pathogenic gliadin fragments and their connection to the early stage of diseases. This review is dedicated to the most relevant biophysical characterization of the complex gliadin digest and the two most studied gliadin fragments, the immunodominant 33-mer peptide and the toxic p31-43 in connection with inflammation and innate immune response. Here, we want to emphasize that combining different biophysical methods with cellular and in vivo models is of key importance to get an integrative understanding of a complex biological problem, as discussed here.
Collapse
|
27
|
Dvoryakova EA, Vinokurov KS, Tereshchenkova VF, Dunaevsky YE, Belozersky MA, Oppert B, Filippova IY, Elpidina EN. Primary digestive cathepsins L of Tribolium castaneum larvae: Proteomic identification, properties, comparison with human lysosomal cathepsin L. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 140:103679. [PMID: 34763092 DOI: 10.1016/j.ibmb.2021.103679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 06/13/2023]
Abstract
We previously described the most highly expressed enzymes from the gut of the red flour beetle, Tribolium castaneum, as cathepsins L. In the present study, two C1 family-specific cysteine cathepsin L enzymes from the larval midgut were isolated and identified using MALDI-TOF MS analysis. The isolated T. castaneum cathepsins were characterized according to their specificity against chromogenic and fluorogenic peptide substrates, and the most efficiently hydrolyzed substrate was Z-FR-pNA with Arg in the P1 subsite. The specificity of insect digestive cathepsins was compared with human lysosomal cathepsin L, the well-studied peptidase of the C1 family cathepsins. T. castaneum digestive cathepsins efficiently hydrolyzed substrates with small and uncharged amino acid residues at P1 (Ala, Gln) more than human cathepsin L. In particular, these insect digestive cathepsins cleaved with higher efficiency the analogs of immunogenic peptides of gliadins, which contribute to autoimmune celiac disease in susceptible people, and thus insect enzymes may be useful in enzymatic treatments for this disease. A bioinformatic study supported by the proteomic analysis of the primary structures of the isolated cathepsins was used to compare tertiary models. The phylogenetic analysis of coleopteran and human cathepsins from the L subfamily indicated that insect digestive cathepsins grouped separately from lysosomal cathepsins.
Collapse
Affiliation(s)
- E A Dvoryakova
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
| | - K S Vinokurov
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Czech Republic, Branišovská 1160/31, České Budějovice, 370 05, Czech Republic
| | - V F Tereshchenkova
- Department of Chemistry, Moscow State University, Moscow, 119991, Russia
| | - Y E Dunaevsky
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
| | - M A Belozersky
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
| | - B Oppert
- USDA Agricultural Research Service, Center for Grain and Animal Health Research, Manhattan, KS, 66502, USA.
| | - I Y Filippova
- Department of Chemistry, Moscow State University, Moscow, 119991, Russia
| | - E N Elpidina
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
28
|
Tanner GJ. Relative Rates of Gluten Digestion by Nine Commercial Dietary Digestive Supplements. Front Nutr 2021; 8:784850. [PMID: 34950690 PMCID: PMC8688929 DOI: 10.3389/fnut.2021.784850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023] Open
Abstract
Endopeptidases containing supplements may digest gluten and reduce the impact on celiac and gluten-sensitive subjects who inadvertently consume gluten. We investigated the relative rate of disappearance of coeliac relevant epitopes in extracts of nine commercial supplements, using two competitive enzyme-linked immunosorbent assays (ELISAs)—Ridascreen (detects QQPFP, QQQFP, LQPFP, and QLPFP) and Gluten-Tec (detects Glia-α20 and PFRPQQPYPQ). All epitopes are destroyed by cleavage after P and Q amino acids. Rates at pH 3.5 and pH 7.0 were measured. These experiments were designed to measure relative rates of epitope digestion not to mimic in vivo digestion. The supplements were: 1 GluteGuard, 2 GlutenBlock, 3 GliadinX, 4 GlutnGo, 5 GlutenRescue, 6 Eat E-Z Gluten+, 7 Glutenease, 8 Glutezyme, and 9 Gluten Digest. The mean initial rate and half-lives of epitope digestion were deduced and extrapolated to rates at the recommended dose of one supplement in a fasting stomach volume. At pH 7, supplement 1 was the fastest acting of the supplements, with Ridascreen ELISA, more than twice as fast as the next fastest supplements, 5, 6, 7, and 8. Supplements 2, 3, and 4 showed little activity at pH 7.0. Supplement 1 was also the fastest acting at pH 7 with Gluten-Tec ELISA, more than three times the rate for supplements 2 and 3, with supplements 4–9 showing minimal activity. At pH 3.5, supplement 1 acted more than five times as fast as the next fastest supplements, 2 and 3, when measured by Ridascreen, but supplements 2 and 3 were over two times faster than supplement 1 when measured by Gluten-Tec. Supplements 4–9 demonstrated minimal activity at pH 3.5 with either ELISA. Supplement 1 most rapidly digested the key immuno-reactive gluten epitopes identified by the R5 antibody in the Codex-approved competitive Ridascreen ELISA method and associated with the pathology of celiac disease.
Collapse
|
29
|
Wu X, Qian L, Liu K, Wu J, Shan Z. Gastrointestinal microbiome and gluten in celiac disease. Ann Med 2021; 53:1797-1805. [PMID: 34647492 PMCID: PMC8519548 DOI: 10.1080/07853890.2021.1990392] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/30/2021] [Indexed: 01/11/2023] Open
Abstract
Coeliac disease (CD), also known as gluten sensitive enteropathy, is an autoimmune intestinal disease induced by gluten in genetically susceptible individuals. Gluten is a common ingredient in daily diet and is one of the main environmental factors to induce coeliac disease. Adhering to gluten free diet (GFD) is an effective method for treating CD. Microbiota plays an extremely important role in maintaining human health, and diet is the main factor to regulate the composition and function of gut microbiota. Recent studies have shown that gluten metabolism is closely related to gastrointestinal tract (GIT) microbiota. With the increasing prevalence of coeliac disease, there is a need for alternative treatments to GFD. In this review, biological medication of gluten, relationship between gluten and gut microflora, effect of GFD on GIT microflora, and effect of probiotics on CD were reviewed. By analysing the research progress on relationship between gluten and gastrointestinal microbiome in coeliac disease, this review tried to explore the prospective and potential mechanism of microecological agents in treating coeliac disease.
Collapse
Affiliation(s)
- Xingxing Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lin Qian
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kexin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Wu
- Institute of Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing University, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaowei Shan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
30
|
Gell G, Bugyi Z, Florides CG, Birinyi Z, Réder D, Szegő Z, Mucsi E, Schall E, Ács K, Langó B, Purgel S, Simon K, Varga B, Vida G, Veisz O, Tömösközi S, Békés F. Investigation of Protein and Epitope Characteristics of Oats and Its Implications for Celiac Disease. Front Nutr 2021; 8:702352. [PMID: 34660657 PMCID: PMC8511309 DOI: 10.3389/fnut.2021.702352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
The use of pure oats (oats cultivated with special care to avoid gluten contamination from wheat, rye, and barley) in the gluten-free diet (GFD) represents important nutritional benefits for the celiac consumer. However, emerging evidence suggests that some oat cultivars may contain wheat gliadin analog polypeptides. Consequently, it is necessary to screen oats in terms of protein and epitope composition to be able to select safe varieties for gluten-free applications. The overall aim of our study is to investigate the variability of oat protein composition directly related to health-related and techno-functional properties. Elements of an oat sample population representing 162 cultivated varieties from 20 countries and the protein composition of resulting samples have been characterized. Size distribution of the total protein extracts has been analyzed by size exclusion-high performance liquid chromatography (SE-HPLC) while the 70% ethanol-extracted proteins were analyzed by RP-HPLC. Protein extracts separated into three main groups of fractions on the SE-HPLC column: polymeric proteins, avenins (both containing three subgroups based on their size), and soluble proteins, representing respectively 68.79–86.60, 8.86–27.72, and 2.89–11.85% of the total protein content. The ratio of polymeric to monomeric proteins varied between 1.37 and 3.73. Seventy-six reversed phase-HPLC-separated peaks have been differentiated from the ethanol extractable proteins of the entire population. Their distribution among the cultivars varied significantly, 6–23 peaks per cultivar. The number of appearances of peaks also showed large variation: one peak has been found in 107 samples, while 15 peaks have been identified, which appeared in less than five cultivars. An estimation method for ranking the avenin-epitope content of the samples has been developed by using MS spectrometric data of collected RP-HPLC peaks and bioinformatics methods. Using ELISA methodology with the R5 antibody, a high number of the investigated samples were found to be contaminated with wheat, barley, or rye.
Collapse
Affiliation(s)
- Gyöngyvér Gell
- Department of Biological Resources, Agricultural Institute, Centre for Agricultural Research, EötvösLoránd Research Network, Martonvásár, Hungary.,Department of Applied Biotechnology and Food Science, Research Group of Cereal Science and Food Quality, Budapest University of Technology and Economics, Budapest, Hungary
| | - Zsuzsanna Bugyi
- Department of Applied Biotechnology and Food Science, Research Group of Cereal Science and Food Quality, Budapest University of Technology and Economics, Budapest, Hungary
| | | | - Zsófia Birinyi
- Department of Biological Resources, Agricultural Institute, Centre for Agricultural Research, EötvösLoránd Research Network, Martonvásár, Hungary
| | - Dalma Réder
- Department of Biological Resources, Agricultural Institute, Centre for Agricultural Research, EötvösLoránd Research Network, Martonvásár, Hungary
| | - Zsuzsanna Szegő
- Department of Applied Biotechnology and Food Science, Research Group of Cereal Science and Food Quality, Budapest University of Technology and Economics, Budapest, Hungary
| | - Edina Mucsi
- Department of Applied Biotechnology and Food Science, Research Group of Cereal Science and Food Quality, Budapest University of Technology and Economics, Budapest, Hungary
| | - Eszter Schall
- Department of Applied Biotechnology and Food Science, Research Group of Cereal Science and Food Quality, Budapest University of Technology and Economics, Budapest, Hungary
| | - Katalin Ács
- Cereal Research Non-Profit Ltd., Szeged, Hungary
| | | | | | | | - Balázs Varga
- Cereal Breeding Department, Agricultural Institute, Centre for Agricultural Research, EötvösLoránd Research Network, Martonvásár, Hungary
| | - Gyula Vida
- Cereal Breeding Department, Agricultural Institute, Centre for Agricultural Research, EötvösLoránd Research Network, Martonvásár, Hungary
| | - Ottó Veisz
- Cereal Breeding Department, Agricultural Institute, Centre for Agricultural Research, EötvösLoránd Research Network, Martonvásár, Hungary
| | - Sándor Tömösközi
- Department of Applied Biotechnology and Food Science, Research Group of Cereal Science and Food Quality, Budapest University of Technology and Economics, Budapest, Hungary
| | | |
Collapse
|
31
|
Dunaevsky YE, Tereshchenkova VF, Belozersky MA, Filippova IY, Oppert B, Elpidina EN. Effective Degradation of Gluten and Its Fragments by Gluten-Specific Peptidases: A Review on Application for the Treatment of Patients with Gluten Sensitivity. Pharmaceutics 2021; 13:1603. [PMID: 34683896 PMCID: PMC8541236 DOI: 10.3390/pharmaceutics13101603] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/22/2021] [Accepted: 09/29/2021] [Indexed: 12/21/2022] Open
Abstract
To date, there is no effective treatment for celiac disease (CD, gluten enteropathy), an autoimmune disease caused by gluten-containing food. Celiac patients are supported by a strict gluten-free diet (GFD). However, in some cases GFD does not negate gluten-induced symptoms. Many patients with CD, despite following such a diet, retain symptoms of active disease due to high sensitivity even to traces of gluten. In addition, strict adherence to GFD reduces the quality of life of patients, as often it is difficult to maintain in a professional or social environment. Various pharmacological treatments are being developed to complement GFD. One promising treatment is enzyme therapy, involving the intake of peptidases with food to digest immunogenic gluten peptides that are resistant to hydrolysis due to a high prevalence of proline and glutamine amino acids. This narrative review considers the features of the main proline/glutamine-rich proteins of cereals and the conditions that cause the symptoms of CD. In addition, we evaluate information about peptidases from various sources that can effectively break down these proteins and their immunogenic peptides, and analyze data on their activity and preliminary clinical trials. Thus far, the data suggest that enzyme therapy alone is not sufficient for the treatment of CD but can be used as a pharmacological supplement to GFD.
Collapse
Affiliation(s)
- Yakov E. Dunaevsky
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (Y.E.D.); (M.A.B.); (E.N.E.)
| | | | - Mikhail A. Belozersky
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (Y.E.D.); (M.A.B.); (E.N.E.)
| | - Irina Y. Filippova
- Chemical Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.F.T.); (I.Y.F.)
| | - Brenda Oppert
- USDA Agricultural Research Service, Center for Grain and Animal Health Research, Manhattan, KS 66502, USA
| | - Elena N. Elpidina
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (Y.E.D.); (M.A.B.); (E.N.E.)
| |
Collapse
|
32
|
Ranking of immunodominant epitopes in celiac disease: Identification of reliable parameters for the safety assessment of innovative food proteins. Food Chem Toxicol 2021; 157:112584. [PMID: 34582965 DOI: 10.1016/j.fct.2021.112584] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/30/2021] [Accepted: 09/22/2021] [Indexed: 01/25/2023]
Abstract
A ranking of gluten T-cell epitopes triggering celiac disease (CD) for its potential application in the safety assessment of innovative food proteins is developed. This ranking takes into account clinical relevance and information derived from key steps involved in the CD pathogenic pathway: enzymatic digestion, epitope binding to HLA-DQ receptors of the antigen-presenting cells and activation of pro-inflammatory CD4 T-cells, which recognizes the HLA-DQ-epitope complex and initiates the inflammatory response. In silico chymotrypsin digestion was the most discriminatory tool for the ranking of gluten T-cell epitopes among all digestive enzymes studied, classifying 81% and 60% of epitopes binding HLA-DQ2.5 and HLA-DQ8 molecules, respectively, with a high risk. A positive relationship between the number of prolines and the risk of gluten T-cell epitopes was identified. HLA-binding data analysis revealed the additional role played by the flanking regions of the 9-mer epitopes whereas the integration of T-cell activation data into the ranking strategy was incomplete because it was difficult to combine results from different studies. The overall ranking proposed in decreasing order of immunological relevance was: α-gliadins > ω-gliadins > hordeins > γ-gliadins ∼ avenins ∼ secalins > glutenins. This novel approach can be considered as a first step to reshape the risk assessment strategy of innovative proteins and their potential to trigger CD.
Collapse
|
33
|
Ribeiro M, de Sousa T, Sabença C, Poeta P, Bagulho AS, 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. [PMID: 34402581 DOI: 10.1111/1541-4337.12828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 05/18/2021] [Accepted: 07/21/2021] [Indexed: 12/21/2022]
Abstract
Gluten-free products have emerged in response to the increasing prevalence of gluten-related disorders, namely celiac disease. Therefore, the quantification of gluten in products intended for consumption by individuals who may suffer from these pathologies must be accurate and reproducible, in a way that allows their proper labeling and protects the health of consumers. Immunochemical methods have been the methods of choice for quantifying gluten, and several kits are commercially available. Nevertheless, they still face problems such as the initial extraction of gluten in complex matrices or the use of a standardized reference material to validate the results. Lately, other methodologies relying mostly on mass spectrometry-based techniques have been explored, and that may allow, in addition to quantitative analysis, the characterizationof gluten proteins. On the other hand, although the level of 20 mg/kg of gluten detected by these methods is sufficient for a product to be considered gluten-free, its immunogenic potential for celiac patients has not been clinically validated. In this sense, in vitro and in vivo models, such as the organoid technology applied in gut-on-chip devices and the transgenic humanized mouse models, respectively, are being developed for investigating both the gluten-induced pathogenesis and the treatment of celiac disease. Due to the ubiquitous nature of gluten in the food industry, as well as the increased prevalence of gluten-related disorders, here we intend to summarize the available methods for gluten quantification in food matrices and for the evaluation of its immunogenic potential concerning the development of novel therapies for celiac disease to highlight active research and discuss knowledge gaps and current challenges in this field.
Collapse
Affiliation(s)
- Miguel Ribeiro
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unity, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, Caparica, Lisbon, Portugal
| | - Telma de Sousa
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unity, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, Caparica, Lisbon, Portugal
| | - Carolina Sabença
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unity, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, Caparica, Lisbon, Portugal
| | - Patrícia Poeta
- LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, Caparica, Lisbon, Portugal.,Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Ana Sofia Bagulho
- National Institute for Agrarian and Veterinarian Research, Elvas, Portugal
| | - Gilberto Igrejas
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unity, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,LAQV-REQUIMTE, Faculty of Science and Technology, University Nova of Lisbon, Caparica, Lisbon, Portugal
| |
Collapse
|
34
|
Herrera MG, Nicoletti F, Gras M, Dörfler PW, Tonali N, Hannappel Y, Ennen I, Hütten A, Hellweg T, Lammers KM, Dodero VI. Pepsin Digest of Gliadin Forms Spontaneously Amyloid-Like Nanostructures Influencing the Expression of Selected Pro-Inflammatory, Chemoattractant, and Apoptotic Genes in Caco-2 Cells: Implications for Gluten-Related Disorders. Mol Nutr Food Res 2021; 65:e2100200. [PMID: 34110092 DOI: 10.1002/mnfr.202100200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/05/2021] [Indexed: 12/11/2022]
Abstract
SCOPE Proteolysis-resistant gliadin peptides are intensely investigated in biomedical research relates to celiac disease and gluten-related disorders. Herein, the first integrated supramolecular investigation of pepsin-digested gliadin peptides (p-gliadin) is presented in combination with its functional behavior in the Caco-2 cell line. METHODS AND RESULTS First, gliadins are degraded by pepsin at pH 3, and the physicochemical properties of p-gliadin are compared with gliadin. An integrated approach using interfacial, spectroscopic, and microscopic techniques reveals that the p-gliadin forms spontaneously soluble large supramolecular structures, mainly oligomers and fibrils, capable of binding amyloid-sensitive dyes. The self-assembly of p-gliadin starts at a concentration of 0.40 µg mL-1 . Second, the stimulation of Caco-2 cells with the p-gliadin supramolecular system is performed, and the mRNA expression levels of a panel of genes are tested. The experiments show that p-gliadin composed of supramolecular structures triggers significant mRNA up-regulation (p < 0.05) of pro-apoptotic biomarkers (ratio Bcl2/Bak-1), chemokines (CCL2, CCL3, CCL4, CCL5, CXCL8), and the chemokine receptor CXCR3. CONCLUSIONS This work demonstrates that p-gliadin is interfacial active, forming spontaneously amyloid-type structures that trigger genes in the Caco-2 cell line involved in recruiting specialized immune cells.
Collapse
Affiliation(s)
- Maria Georgina Herrera
- Department of Chemistry, Organic Chemistry III, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany.,Institute of Biological Chemistry and Physical Chemistry, Dr. Alejandro Paladini, University of Buenos Aires-CONICET, Buenos Aires, C1113AAD, Argentina
| | - Francesco Nicoletti
- Department of Chemistry, Organic Chemistry III, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany
| | - Marion Gras
- Department of Chemistry, Organic Chemistry III, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany.,Department of Chemistry, Physical and Biophysical Chemistry, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany
| | - Philipp W Dörfler
- Department of Chemistry, Organic Chemistry III, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany
| | - Nicolo Tonali
- Department of Chemistry, Organic Chemistry III, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany.,Faculté de Pharmacie, Université Paris-Saclay, BioCIS, 5 rue Jean-Baptiste Clément, Châtenay-Malabry, 92296, France
| | - Yvonne Hannappel
- Department of Chemistry, Physical and Biophysical Chemistry, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany
| | - Inga Ennen
- Department of Physics, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany
| | - Andreas Hütten
- Department of Physics, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany
| | - Thomas Hellweg
- Department of Chemistry, Physical and Biophysical Chemistry, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany
| | - Karen M Lammers
- Department of Chemistry, Organic Chemistry III, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany.,Tubascan Ltd., Science Park 106, Amsterdam, 1098 XG, the Netherlands
| | - Veronica I Dodero
- Department of Chemistry, Organic Chemistry III, Bielefeld University, Universitätsstr. 25, Bielefeld, 33615, Germany
| |
Collapse
|
35
|
A Case Study of the Response of Immunogenic Gluten Peptides to Sourdough Proteolysis. Nutrients 2021; 13:nu13061906. [PMID: 34206002 PMCID: PMC8229354 DOI: 10.3390/nu13061906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Celiac disease is activated by digestion-resistant gluten peptides that contain immunogenic epitopes. Sourdough fermentation is a potential strategy to reduce the concentration of these peptides within food. However, we currently know little about the effect of partial sourdough fermentation on immunogenic gluten. This study examined the effect of a single sourdough culture (representative of those that the public may consume) on the digestion of immunogenic gluten peptides. Sourdough bread was digested via the INFOGEST protocol. Throughout digestion, quantitative and discovery mass spectrometry were used to model the kinetic release profile of key immunogenic peptides and profile novel peptides, while ELISA probed the gluten's allergenicity. Macrostructural studies were also undertaken. Sourdough fermentation altered the protein structure, in vitro digestibility, and immunogenic peptide release profile. Interestingly, sourdough fermentation did not decrease the total immunogenic peptide concentration but altered the in vitro digestion profile of select immunogenic peptides. This work demonstrates that partial sourdough fermentation can alter immunogenic gluten digestion, and is the first study to examine the in vitro kinetic profile of immunogenic gluten peptides from sourdough bread.
Collapse
|
36
|
Diós Á, Elek R, Szabó I, Horváth S, Gyimesi J, Király R, Werkstetter K, Koletzko S, Fésüs L, Korponay-Szabó IR. Gamma-gliadin specific celiac disease antibodies recognize p31-43 and p57-68 alpha gliadin peptides in deamidation related manner as a result of cross-reaction. Amino Acids 2021; 53:1051-1063. [PMID: 34059947 PMCID: PMC8241804 DOI: 10.1007/s00726-021-03006-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 05/17/2021] [Indexed: 12/02/2022]
Abstract
Celiac disease (CeD) is a T-cell-dependent enteropathy with autoimmune features where tissue transglutaminase (TG2)-mediated posttranslational modification of gliadin peptides has a decisive role in the pathomechanism. The humoral immune response is reported to target mainly TG2-deamidated γ-gliadin peptides. However, α-gliadin peptides, like p57-68, playing a crucial role in the T-cell response, and p31-43, a major trigger of innate responses, also contain B-cell gliadin epitopes and γ-gliadin like motifs. We aimed to identify if there are anti-gliadin-specific antibodies in CeD patients targeting the p31-43 and p57-68 peptides and to examine whether deamidation of these peptides could increase their antigenicity. We explored TG2-mediated deamidation of the p31-43 and p57-68 peptides, and investigated serum antibody reactivity toward the native and deamidated α and γ-gliadin peptides in children with confirmed CeD and in prospectively followed infants at increased risk for developing CeD. We affinity-purified antibody populations utilizing different single peptide gliadin antigens and tested their binding preferences for cross-reactivity in real-time interaction assays based on bio-layer interferometry. Our results demonstrate that there is serum reactivity toward p31-43 and p57-68 peptides, which is due to cross-reactive γ-gliadin specific antibodies. These γ-gliadin specific antibodies represent the first appearing antibody population in infancy and they dominate the serum reactivity of CeD patients even later on and without preference for deamidation. However, for the homologous epitope sequences in α-gliadins shorter than the core QPEQPFP heptapeptide, deamidation facilitates antibody recognition. These findings reveal the presence of cross-reactive antibodies in CeD patients recognizing the disease-relevant α-gliadins.
Collapse
Affiliation(s)
- Ádám Diós
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Rita Elek
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildikó Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Horváth
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Gyimesi
- Coeliac Disease Center, Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Róbert Király
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katharina Werkstetter
- Division of Gastroenterology and Hepatology, Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilian's University Munich, Munich, Germany
| | - Sibylle Koletzko
- Division of Gastroenterology and Hepatology, Department of Pediatrics, Dr. Von Hauner Children's Hospital, University Hospital, Ludwig-Maximilian's University Munich, Munich, Germany.,Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ilma R Korponay-Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary. .,Coeliac Disease Center, Heim Pál National Pediatric Institute, Budapest, Hungary.
| |
Collapse
|
37
|
Woldemariam KY, Yuan J, Wan Z, Yu Q, Cao Y, Mao H, Liu Y, Wang J, Li H, Sun B. Celiac Disease and Immunogenic Wheat Gluten Peptides and the Association of Gliadin Peptides with HLA DQ2 and HLA DQ8. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2021.1907755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Kalekristos Yohannes Woldemariam
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| | - Juanli Yuan
- School of Pharmacy, Nanchang University, Nanchang, China
| | - Zhen Wan
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| | - Qinglin Yu
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| | - Yating Cao
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| | - Huijia Mao
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| | - Yingli Liu
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| | - Jing Wang
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| | - Hongyan Li
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| | - Baoguo Sun
- School of Food and Health, China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU), Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing, China
| |
Collapse
|
38
|
Kurki A, Kemppainen E, Laurikka P, Kaukinen K, Lindfors K. The use of peripheral blood mononuclear cells in celiac disease diagnosis and treatment. Expert Rev Gastroenterol Hepatol 2021; 15:305-316. [PMID: 33176106 DOI: 10.1080/17474124.2021.1850262] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Celiac disease is characterized by an abnormal immune activation driven by the ingestion of gluten from wheat, barley, and rye. Gluten-specific CD4+ T cells play an important role in disease pathogenesis and are detectable among peripheral blood mononuclear cells (PBMCs). Areas covered: This review summarizes the use of celiac disease patient PBMCs in clinical applications focusing on their exploitation in the development of diagnostic approaches and novel drugs to replace or complement gluten-free diet. Expert opinion: The most used PBMC-based methods applied in celiac disease research include ELISpot and HLA-DQ:gluten tetramer technology. ELISpot has been utilized particularly in research aiming to develop a celiac disease vaccine and in studies addressing the toxicity of different grains in celiac disease. HLA-DQ:gluten tetramer technology on the other hand initially focused on improving current diagnostics but in combination with additional markers it is also a useful outcome measure in clinical trials to monitor the efficacy of drug candidates. In addition, the technology serves well in the more detailed characterization of celiac disease-specific T cells, thereby possibly revealing novel therapeutic targets. Future studies may also reveal clinical applications for PBMC microRNAs and/or dendritic cells or monocytes present among PBMCs.
Collapse
Affiliation(s)
- Alma Kurki
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University , Tampere, Finland
| | - Esko Kemppainen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University , Tampere, Finland
| | - Pilvi Laurikka
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University , Tampere, Finland
| | - Katri Kaukinen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University , Tampere, Finland.,Department of Internal Medicine, Tampere University Hospital , Tampere, Finland
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University , Tampere, Finland
| |
Collapse
|
39
|
The effect of baking time and temperature on gluten protein structure and celiac peptide digestibility. Food Res Int 2021; 140:109988. [DOI: 10.1016/j.foodres.2020.109988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/20/2022]
|
40
|
Gliadin Sequestration as a Novel Therapy for Celiac Disease: A Prospective Application for Polyphenols. Int J Mol Sci 2021; 22:ijms22020595. [PMID: 33435615 PMCID: PMC7826989 DOI: 10.3390/ijms22020595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
Celiac disease is an autoimmune disorder characterized by a heightened immune response to gluten proteins in the diet, leading to gastrointestinal symptoms and mucosal damage localized to the small intestine. Despite its prevalence, the only treatment currently available for celiac disease is complete avoidance of gluten proteins in the diet. Ongoing clinical trials have focused on targeting the immune response or gluten proteins through methods such as immunosuppression, enhanced protein degradation and protein sequestration. Recent studies suggest that polyphenols may elicit protective effects within the celiac disease milieu by disrupting the enzymatic hydrolysis of gluten proteins, sequestering gluten proteins from recognition by critical receptors in pathogenesis and exerting anti-inflammatory effects on the system as a whole. This review highlights mechanisms by which polyphenols can protect against celiac disease, takes a critical look at recent works and outlines future applications for this potential treatment method.
Collapse
|
41
|
Filippova IY, Dvoryakova EA, Sokolenko NI, Simonyan TR, Tereshchenkova VF, Zhiganov NI, Dunaevsky YE, Belozersky MA, Oppert B, Elpidina EN. New Glutamine-Containing Substrates for the Assay of Cysteine Peptidases From the C1 Papain Family. Front Mol Biosci 2020; 7:578758. [PMID: 33195423 PMCID: PMC7643032 DOI: 10.3389/fmolb.2020.578758] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/07/2020] [Indexed: 01/06/2023] Open
Abstract
New substrates with glutamine in the P1-position are introduced for the assay of peptidases from the C1 papain family, with a general formula of Glp-Phe-Gln-X, where Glp is pyroglutamyl and X is pNA (p-nitroanilide) or AMC (4-amino-7-methylcoumaride). The substrates have a simple structure, and C1 cysteine peptidases of various origins cleave them with high efficiency. The main advantage of the substrates is their selectivity for cysteine peptidases of the C1 family. Peptidases of other clans, including serine trypsin-like peptidases, do not cleave glutamine-containing substrates. We demonstrate that using Glp-Phe-Gln-pNA in combination with a commercially available substrate, Z-Arg-Arg-pNA, provided differential determination of cathepsins L and B. In terms of specific activity and kinetic parameters, the proposed substrates offer improvement over the previously described alanine-containing prototypes. The efficiency and selectivity of the substrates was demonstrated by the example of chromatographic and electrophoretic analysis of a multi-enzyme digestive complex of stored product pests from the Tenebrionidae family.
Collapse
Affiliation(s)
- Irina Y Filippova
- Division of Natural Compounds, Department of Chemistry, Moscow State University, Moscow, Russia
| | - Elena A Dvoryakova
- Department of Plant Proteins, A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - Nikolay I Sokolenko
- Laboratory of Protein Chemistry, Institute of Genetics and Selection of Industrial Microorganisms, Moscow, Russia
| | - Tatiana R Simonyan
- Division of Natural Compounds, Department of Chemistry, Moscow State University, Moscow, Russia
| | | | - Nikita I Zhiganov
- Division of Entomology, Faculty of Biology, Moscow State University, Moscow, Russia
| | - Yakov E Dunaevsky
- Department of Plant Proteins, A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - Mikhail A Belozersky
- Department of Plant Proteins, A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - Brenda Oppert
- USDA Agricultural Research Service, Center for Grain and Animal Health Research, Manhattan, KS, United States
| | - Elena N Elpidina
- Department of Plant Proteins, A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| |
Collapse
|
42
|
Pronin D, Börner A, Scherf KA. Old and modern wheat (Triticum aestivum L.) cultivars and their potential to elicit celiac disease. Food Chem 2020; 339:127952. [PMID: 33152854 DOI: 10.1016/j.foodchem.2020.127952] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/19/2022]
Abstract
One potential explanation for the increasing prevalence of celiac disease (CD) over the past decades is that breeding may have inadvertently changed the immunoreactive potential of wheat. To test this hypothesis, we quantitated four CD-active peptides, namely the 33-mer and peptides containing the DQ2.5-glia-α1a/DQ2.5-glia-α2 (P1), DQ2.5-glia-α3 (P2) and DQ2.5-glia-γ1 (P3) epitopes, in a set of 60 German hexaploid winter wheat cultivars from 1891 to 2010 and grown in three consecutive years. The contents of CD-active peptides were affected more by the harvest year than by the cultivar. The 33-mer and P1 peptides showed no tendency regarding their absolute contents in the flour, but they tended to increase slightly over time when calculated relative to the α-gliadins. No trends in relative or absolute values were observed for the P2 and P3 peptides derived from α- and γ-gliadins. Therefore, the immunoreactive potential of old and modern wheat cultivars appears to be similar.
Collapse
Affiliation(s)
- Darina Pronin
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany.
| | - Andreas Börner
- Genebank Department, Leibniz Institute of Plant Genetics and Crop Plant Research, Corrensstr. 3, 06466 Seeland, OT Gatersleben, Germany.
| | - Katharina Anne Scherf
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; Department of Bioactive and Functional Food Chemistry, Institute of Applied Biosciences, Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131 Karlsruhe, Germany.
| |
Collapse
|
43
|
Microbiome of root vegetables-a source of gluten-degrading bacteria. Appl Microbiol Biotechnol 2020; 104:8871-8885. [PMID: 32875365 PMCID: PMC7502452 DOI: 10.1007/s00253-020-10852-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/29/2020] [Accepted: 08/23/2020] [Indexed: 12/11/2022]
Abstract
Abstract Gluten is a cereal protein that is incompletely digested by human proteolytic enzymes that create immunogenic peptides that accumulate in the gastrointestinal tract (GIT). Although both environmental and human bacteria have been shown to expedite gluten hydrolysis, gluten intolerance is a growing concern. Here we hypothesize that together with food, we acquire environmental bacteria that could impact our GIT with gluten-degrading bacteria. Using in vitro gastrointestinal simulation conditions, we evaluated the capacity of endophytic bacteria that inhabit root vegetables, potato (Solanum tuberosum), carrot (Daucus sativus), beet (Beta vulgaris), and topinambur (Jerusalem artichoke) (Helianthus tuberosus), to resist these conditions and degrade gluten. By 16S rDNA sequencing, we discovered that bacteria from the families Enterobacteriaceae, Bacillaceae, and Clostridiaceae most effectively multiply in conditions similar to the human GIT (microoxic conditions, 37 °C) while utilizing vegetable material and gluten as nutrients. Additionally, we used stomach simulation (1 h, pH 3) and intestinal simulation (1 h, bile salts 0.4%) treatments. The bacteria that survived this treatment retained the ability to degrade gluten epitopes but at lower levels. Four bacterial strains belonging to species Bacillus pumilus, Clostridium subterminale, and Clostridium sporogenes isolated from vegetable roots produced proteases with postproline cleaving activity that successfully neutralized the toxic immunogenic epitopes. Key points • Bacteria from root vegetables can degrade gluten. • Some of these bacteria can resist conditions mimicking gastrointestinal tract. Electronic supplementary material The online version of this article (10.1007/s00253-020-10852-0) contains supplementary material, which is available to authorized users.
Collapse
|
44
|
Dunaevsky YE, Tereshchenkova VF, Oppert B, Belozersky MA, Filippova IY, Elpidina EN. Human proline specific peptidases: A comprehensive analysis. Biochim Biophys Acta Gen Subj 2020; 1864:129636. [DOI: 10.1016/j.bbagen.2020.129636] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/05/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
|
45
|
Gluten Degrading Enzymes for Treatment of Celiac Disease. Nutrients 2020; 12:nu12072095. [PMID: 32679754 PMCID: PMC7400306 DOI: 10.3390/nu12072095] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Celiac disease (CeD) affects about 1% of most world populations. It presents a wide spectrum of clinical manifestations, ranging from minor symptoms to mild or severe malabsorption, and it may be associated with a wide variety of autoimmune diseases. CeD is triggered and maintained by the ingestion of gluten proteins from wheat and related grains. Gluten peptides that resist gastrointestinal digestion are antigenically presented to gluten specific T cells in the intestinal mucosa via HLA-DQ2 or HLA-DQ8, the necessary genetic predisposition for CeD. To date, there is no effective or approved treatment for CeD other than a strict adherence to a gluten-free diet, which is difficult to maintain in professional or social environments. Moreover, many patients with CeD have active disease despite diet adherence due to a high sensitivity to traces of gluten. Therefore, safe pharmacological treatments that complement the gluten-free diet are urgently needed. Oral enzyme therapy, employing gluten-degrading enzymes, is a promising therapeutic approach. A prerequisite is that such enzymes are active under gastro-duodenal conditions, quickly neutralize the T cell activating gluten peptides and are safe for human consumption. Several enzymes including prolyl endopeptidases, cysteine proteases and subtilisins can cleave the human digestion-resistant gluten peptides in vitro and in vivo. Examples are several prolyl endopeptidases from bacterial sources, subtilisins from Rothia bacteria that are natural oral colonizers and synthetic enzymes with optimized gluten-degrading activities. Without exception, these enzymes must cleave the otherwise unusual glutamine and proline-rich domains characteristic of antigenic gluten peptides. Moreover, they should be stable and active in both the acidic environment of the stomach and under near neutral pH in the duodenum. This review focuses on those enzymes that have been characterized and evaluated for the treatment of CeD, discussing their origin and activities, their clinical evaluation and challenges for therapeutic application. Novel developments include strategies like enteric coating and genetic modification to increase enzyme stability in the digestive tract.
Collapse
|
46
|
Rashmi BS, Gayathri D, Vasudha M, Prashantkumar CS, Swamy CT, Sunil KS, Somaraja PK, Prakash P. Gluten hydrolyzing activity of Bacillus spp isolated from sourdough. Microb Cell Fact 2020; 19:130. [PMID: 32532261 PMCID: PMC7291523 DOI: 10.1186/s12934-020-01388-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 06/06/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Celiac disease is an intestinal chronic disorder with multifactorial etiology resulting in small intestinal mucosal injuries and malabsorption. In genetically predisposed individuals with HLA DQ2/DQ8 molecules, the gluten domains rich in glutamine and proline present gluten domains to gluten reactive CD4+ T cells causing injury to the intestine. In the present experimental design, the indigenous bacteria from wheat samples were studied for their gluten hydrolyzing functionality. RESULTS Proteolytic activity of Bacillus spp. was confirmed spectrophotometrically and studied extensively on gliadin-derived synthetic enzymatic substrates, natural gliadin mixture, and synthetic highly immunogenic 33-mer peptide. The degradation of 33-mer peptide and the cleavage specificities of the selected isolates were analyzed by tandem mass spectrometry. The gluten content of the sourdough fermented by the chosen bacterial isolates was determined by R5 antibody based competitive ELISA. All the tested isolates efficiently hydrolyzed Z-YPQ-pNA, Z-QQP-pNA, Z-PPF-pNA, and Z-PFP-pNA and also cleaved 33-mer immunogenic peptide extensively. The gluten content of wheat sourdough was found to be below 110 mg/kg. CONCLUSION It has been inferred that four Bacillus spp especially GS 188 could be useful in developing gluten-reduced wheat food product for celiac disease prone individuals.
Collapse
Affiliation(s)
| | - Devaraja Gayathri
- Department of Microbiology, Davangere University, Davangere, Karnataka, 577002, India.
| | - Mahanthesh Vasudha
- Department of Microbiology, Davangere University, Davangere, Karnataka, 577002, India
| | | | | | - Kumar S Sunil
- Department of Microbiology, Davangere University, Davangere, Karnataka, 577002, India
| | | | - Patil Prakash
- SDM Research Institute for Biomedical Sciences (SDMRIBS), Shri Dharmasthala Manjunatheshwara University, Manjushree Nagar, Dharwad, Karnataka, 580 009, India
- Central Research Laboratory, K S Hegde Medical Academy, Deralakatte, Mangaluru, 575018, India
| |
Collapse
|
47
|
Picascia S, Camarca A, Malamisura M, Mandile R, Galatola M, Cielo D, Gazza L, Mamone G, Auricchio S, Troncone R, Greco L, Auricchio R, Gianfrani C. 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. [PMID: 32374905 DOI: 10.1002/mnfr.201901032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/11/2020] [Indexed: 12/27/2022]
Abstract
SCOPE Gluten from the diploid wheat Triticum monococcum (TM) has low content of immunostimulatory sequences and a high gastro-intestinal digestibility. Gluten-reactive T cells elicited by diploid and hexaploid (Triticum aestivum-TA) wheat in celiac disease (CD) patients upon a brief oral challenge are analyzed. METHODS AND RESULTS Seventeen patients with CD (median age 13 years) consumed for 3 days sandwiches made with TM (cultivar Norberto-ID331, N=11), or TA (cultivar Sagittario, N=11) flours, corresponding to 12 gr of gluten/die. Immunostimulatory properties are assessed in blood by measuring the IFN-γ-secreting T cells by EliSpot and the expression of inflammatory cytokines/receptors (IL-12A, IL-15, IL-18RAP, IFN-γ) by qPCR. TA mobilizes a remarkable number of gliadin-specific, IFN-γ-secreting T cells (p<0.05), while no significant cell mobilization is induced by TM (p=ns). Similar results are obtained in response to five immunogenic peptides from α-, ω-, and γ-gliadins, although with a large individual variability. An increased mRNA expression for IL-12A and IFN-γ is detected in the group eating TA compared to those consuming TM (p<0.05). CONCLUSIONS Although T. monococcum is a cereal not suitable for the diet of celiacs, this diploid wheat elicits a reduced in vivo T-cell response compared to T. aestivum in celiac patients.
Collapse
Affiliation(s)
- Stefania Picascia
- Institute of Biochemistry and Cell Biology, National Research Council, Via Pietro Castellino, 111, Napoli, Naples, 80131, Italy
| | - Alessandra Camarca
- Institute of Food Sciences, National Research Council, Via Roma, 64, Avellino, 83100, Italy
| | - Monica Malamisura
- Department of Medical Translational Sciences, Section of Pediatrics, Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Roberta Mandile
- Department of Medical Translational Sciences, Section of Pediatrics, Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Martina Galatola
- Department of Medical Translational Sciences, Section of Pediatrics, Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Donatella Cielo
- Department of Medical Translational Sciences, Section of Pediatrics, Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Laura Gazza
- CREA Research Centre for Engineering and Agro-Food Processing, Via Po, 14, Roma, 00198, Italy
| | - Gianfranco Mamone
- Institute of Food Sciences, National Research Council, Via Roma, 64, Avellino, 83100, Italy
| | - Salvatore Auricchio
- Department of Medical Translational Sciences, Section of Pediatrics, Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy.,European Laboratory for the Food Induced Diseases (E.L.F.I.D), Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Riccardo Troncone
- Department of Medical Translational Sciences, Section of Pediatrics, Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy.,European Laboratory for the Food Induced Diseases (E.L.F.I.D), Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Luigi Greco
- Department of Medical Translational Sciences, Section of Pediatrics, Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy.,European Laboratory for the Food Induced Diseases (E.L.F.I.D), Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Renata Auricchio
- Department of Medical Translational Sciences, Section of Pediatrics, Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy.,European Laboratory for the Food Induced Diseases (E.L.F.I.D), Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Carmen Gianfrani
- Institute of Biochemistry and Cell Biology, National Research Council, Via Pietro Castellino, 111, Napoli, Naples, 80131, Italy.,European Laboratory for the Food Induced Diseases (E.L.F.I.D), Federico II University of Naples, Via Sergio Pansini, 5, Napoli, 80131, Italy
| |
Collapse
|
48
|
Lexhaller B, Ludwig C, Scherf KA. Identification of Isopeptides Between Human Tissue Transglutaminase and Wheat, Rye, and Barley Gluten Peptides. Sci Rep 2020; 10:7426. [PMID: 32367038 PMCID: PMC7198585 DOI: 10.1038/s41598-020-64143-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/09/2020] [Indexed: 01/28/2023] Open
Abstract
Celiac disease (CD) is a chronic immune-mediated enteropathy of the small intestine, which is triggered by the ingestion of storage proteins (gluten) from wheat, rye, and barley in genetically predisposed individuals. Human tissue transglutaminase (TG2) plays a central role in the pathogenesis of CD, because it is responsible for specific gluten peptide deamidation and covalent crosslinking, resulting in the formation of Nε-(γ-glutamyl)-lysine isopeptide bonds. The resulting TG2-gluten peptide complexes are assumed to cause the secretion of anti-TG2 autoantibodies, but the underlying mechanisms are only partly known. To gain more insight into the structures of these complexes, the aim of our study was to identify TG2-gluten isopeptides. With the use of discovery-driven as well as targeted nanoscale liquid chromatography tandem mass spectrometry, we detected 29 TG2-gluten isopeptides in total, involving seven selected TG2 lysine residues (K205, K265, K429, K468, K590, K600, K677). Several gluten peptides carried known B-cell epitopes and/or T-cell epitopes, either intact 9-mer core regions or partial sequences, as well as sequences bearing striking similarities to already known epitopes. These novel insights into the molecular structures of TG2-gluten peptide complexes may help clarify their physiological relevance in the initiation of CD autoimmunity and the role of anti-TG2 autoantibodies.
Collapse
Affiliation(s)
- Barbara Lexhaller
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354, Freising, Germany
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich, Gregor-Mendel-Str. 4, 85354, Freising, Germany
| | - Katharina Anne Scherf
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354, Freising, Germany. .,Department of Bioactive and Functional Food Chemistry, Institute of Applied Biosciences, Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131, Karlsruhe, Germany.
| |
Collapse
|
49
|
Hardy MY, Russell AK, Pizzey C, Jones CM, Watson KA, La Gruta NL, Cameron DJ, Tye-Din JA. Characterisation of clinical and immune reactivity to barley and rye ingestion in children with coeliac disease. Gut 2020; 69:830-840. [PMID: 31462555 DOI: 10.1136/gutjnl-2019-319093] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/23/2019] [Accepted: 08/15/2019] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Barley and rye are major components of the Western diet, and historic feeding studies indicate that they cause clinical effects in patients with coeliac disease (CD). This toxicity has been attributed to sequence homology with immunogenic wheat sequences, but in adults with CD, these cereals stimulate unique T cells, indicating a critical contribution to gluten immunity independent of wheat. Clinical and immune feeding studies with these grains in children with CD are sparse. We undertook a barley and rye feeding study to characterise the clinical and T-cell responses in children with CD. DESIGN 42 children with human leucocyte antigen (HLA)-DQ2.5+ (aged 3-17 years) consumed barley or rye for 3 days. Blood-derived gluten-specific T cells were tested for reactivity against a panel of barley (hordein) and rye (secalin) peptides. Hordein and secalin-specific T-cell clones were generated and tested for grain cross-reactivity. T-cell receptor sequencing was performed on sorted single cells. T-cell responses were compared with those observed in adults with CD. RESULTS 90% of the children experienced adverse symptoms, mostly GI, and 61% had detectable gluten-specific T-cell responses targeting peptides homologous to those immunogenic in adults. Deamidation was important for peptide reactivity. Homozygosity for HLA-DQ2.5 predicted a stronger T-cell response. Gluten-specific T cells showed striking similarities in their cross-reactivity between children and adults. CONCLUSIONS Barley and rye induce a consistent range of clinical and T-cell responses in children with CD. The findings highlight the importance of a series of dominant hordein and secalin peptides pathogenic in children with CD, some independent of wheat, which closely correspond to those seen in adults.
Collapse
Affiliation(s)
- Melinda Y Hardy
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Amy K Russell
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Catherine Pizzey
- Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Claerwen M Jones
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Katherine A Watson
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Nicole L La Gruta
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Donald J Cameron
- Department of Gastroenterology and Clinical Nutrition, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia .,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, Victoria, Australia.,Centre for Food and Allergy Research, Murdoch Children's Research Institute, Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
50
|
Bekalu ZE, Dionisio G, Brinch-Pedersen H. Molecular Properties and New Potentials of Plant Nepenthesins. PLANTS (BASEL, SWITZERLAND) 2020; 9:E570. [PMID: 32365700 PMCID: PMC7284499 DOI: 10.3390/plants9050570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022]
Abstract
Nepenthesins are aspartic proteases (APs) categorized under the A1B subfamily. Due to nepenthesin-specific sequence features, the A1B subfamily is also named nepenthesin-type aspartic proteases (NEPs). Nepenthesins are mostly known from the pitcher fluid of the carnivorous plant Nepenthes, where they are availed for the hydrolyzation of insect protein required for the assimilation of insect nitrogen resources. However, nepenthesins are widely distributed within the plant kingdom and play significant roles in plant species other than Nepenthes. Although they have received limited attention when compared to other members of the subfamily, current data indicates that they have exceptional molecular and biochemical properties and new potentials as fungal-resistance genes. In the current review, we provide insights into the current knowledge on the molecular and biochemical properties of plant nepenthesins and highlights that future focus on them may have strong potentials for industrial applications and crop trait improvement.
Collapse
Affiliation(s)
- Zelalem Eshetu Bekalu
- Department of Agroecology, Research Center Flakkebjerg, Aarhus University, DK-4200 Slagelse, Denmark; (G.D.); (H.B.-P.)
| | | | | |
Collapse
|