Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Biochem Biophys Res Commun ; 695: 149439, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38160531

ABSTRACT

Celiac disease and other types of gluten intolerance significantly affect the life quality of patients making them restrict the diet removing all food produced from wheat, rye, oat, and barley flour, and some other products. These disorders arise from protease resistance of poorly soluble proteins prolamins, contained in gluten. Enhanced proteolytic digestion of gliadins might be considered as a prospective approach for the treatment of celiac disease and other types of gluten intolerance. Herein, we tested a range of sulfated polymers (kappa-carrageenan, dextran sulfate and different polysaccharides from brown seaweeds, and a synthetic polystyrene sulfonate) for the ability to activate gliadin digestion by human digestive proteases, pepsin and trypsin. Sulfated polysaccharide from Fucus evanescens enhanced proteolytic digestion of gliadins from wheat flour and reduced its cytotoxicity on intestinal epithelial Caco-2 cell culture. Regarding the non-toxic nature of fucoidans, the results provide a basis for polymer-based drugs or additives for the symptomatic treatment of gluten intolerance.


Subject(s)
Celiac Disease , Gliadin , Humans , Gliadin/toxicity , Gliadin/metabolism , Caco-2 Cells , Flour , Sulfates , Triticum , Glutens/metabolism , Peptide Hydrolases , Polysaccharides/pharmacology , Digestion
2.
Inflammation ; 44(3): 873-889, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33394186

ABSTRACT

Wheat is a major diet from many years; apart from its nutritious value, the wheat protein gliadin is responsible for many inflammatory diseases like celiac disease (CD), and non-celiac gluten sensitivity (NCGS). In this study, the gliadin-induced inflammation and associated cellular damage along with the protective role of curcumin was evaluated using human intestinal cell lines (HCT-116 and HT-29) as a model. Cells were cultured and exposed to 160 µg/ml of gliadin, 100 µM H2O2, and 10 µM curcumin (3 h pretreatment) followed by the assessment of inflammation. Spectrophotometric methods, real-time-PCR, ELISA, Western blotting, and confocal microscopy techniques were used to assess inflammatory markers such as advanced oxidation protein products (AOPPs) level, activity of myeloperoxidase (MPO) and NADPH oxidase (NOX), cytokines, and cell damage markers. The results show that gliadin increases the AOPPs level and the activity of MPO and NOX expression. It enhances inflammation by increasing expression of pro-inflammatory cytokines, altered expression of anti-inflammatory, and regulatory cytokines. It exacerbates the cellular damage by increasing MMP-2 and 9 and decreasing integrin α and ß expression. Gliadin promotes disease pathogenesis by inducing the inflammation and cellular damage which further alter the cellular homeostasis. The pretreatment of curcumin counteracts the adverse effect of gliadin and protect the cells via diminishing the inflammation and help the cell to regain the cellular morphology suggesting phytochemical-based remedial interventions against wheat allergies.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Celiac Disease/prevention & control , Curcumin/pharmacology , Enteritis/prevention & control , Gliadin/toxicity , Inflammation Mediators/metabolism , Intestinal Mucosa/drug effects , Wheat Hypersensitivity/prevention & control , Celiac Disease/genetics , Celiac Disease/metabolism , Celiac Disease/pathology , Cytokines/genetics , Cytokines/metabolism , Enteritis/genetics , Enteritis/metabolism , Enteritis/pathology , HCT116 Cells , HT29 Cells , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Oxidative Stress , Signal Transduction , Wheat Hypersensitivity/genetics , Wheat Hypersensitivity/metabolism , Wheat Hypersensitivity/pathology
3.
J Physiol Pharmacol ; 71(2)2020 Apr.
Article in English | MEDLINE | ID: mdl-32633238

ABSTRACT

Celiac disease (CD) is an autoimmune condition that occurs in genetically predisposed people where the ingestion of gluten produces damage in the small intestine. The treatment accepted until now is a strict gluten free diet. This implies the need for novel or adjuvant treatments, in addition to the standard of care. The present study aimed to assess the effect of gold nanoparticles phytosynthesized with Cornus mas extract (AuCM) compared to Cornus mas extract (CM) and luteolin (LT) on Caco-2 cells, exposed or not to gliadin. Ultraviolet-visible spectroscopy and transmission electron microscopy were used for the characterization of AuCM. Measured cellular outcomes included oxidative stress markers (malondialdehyde level, catalase and superoxide dismutase activities), inflammatory response and cellular signaling and transcription factors involved in apoptosis (NFκB, pNFκB, NOS2, TNF-α, TRAIL, Bax, Bcl-2, p53). The internalization of gold nanoparticles in cells was evidenced by transmission electron microscopy (TEM). The gliadin administration induced oxidative stress, improved the activity of antioxidants enzymes, increased NOS2 and NFκB expressions and reduced pNFκB/NFκB ratio. In addition, gliadin enhanced TRAIL and Bcl-2 levels and reduced p53 expression in Caco-2 cells. The pretreatment with AuCM, CM extract and LT diminished oxidative stress and reduced NOS2 activity. AuCM and CM treatment amplified the expression of p53 and pNFκB/NFκB ratio and diminished Bcl-2, NFκB and pNFκB, especially AuCM. The results obtained confirmed that AuCM mitigate some of gliadin effects on Caco-2 cells through modulation of oxidative stress and inflammation.


Subject(s)
Colonic Neoplasms/drug therapy , Cornus/chemistry , Gliadin/toxicity , Gold/chemistry , Metal Nanoparticles/administration & dosage , Plant Extracts/pharmacology , Antioxidants/metabolism , Apoptosis/drug effects , Caco-2 Cells , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Humans , Inflammation/drug therapy , Metal Nanoparticles/chemistry , Oxidative Stress/drug effects
4.
Food Chem Toxicol ; 140: 111331, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32304812

ABSTRACT

The resemblance of physiological traits of cell lines with their target/original tissue is an important prerequisite for the choice of the in vitro model. Although cytoprotective defenses, activated by the nuclear factor erythroid 2-related factor2 (Nrf2), have a preeminent importance in intestinal protection, nevertheless their levels inin vitro models have been never compared with those of their original tissue. Basal level of Nrf2-mediated defenses in murine enterocyte cells (Mode-K) and in human colon adenocarcinoma cells -at differentiated (DCaco2) or confluent stage (CCaco2)- were compared with those found in mouse or human duodenum. The pro-oxidant and cytotoxic effects of peptic-tryptic digest of gluten prepared from wheat bread (PT-b), einkorn (PT-e) or durum wheat (PT-d) were evaluated in Mode-k and DCaco2 by combining enzymatic, immune-enzymatic and real-time PCR assay. The results presented reveal that Mode-k cells resemble cytoprotective defenses of human/murine duodenum and are more susceptible to pro-oxidant, cytotoxic and pro-inflammatory effect of gliadin digest (in comparison with Caco2). Prolamins digests from the considered wheat exhibit different inhibitory effect on Nrf2-mediated defenses (PT-b > PT-e > PT-d). These data indicate, for the first time, that Mode-k are a reliable model for investigating wheat prolamins toxicity and for evaluating the signaling pathway of gluten-associated disease.


Subject(s)
Enterocytes/drug effects , Glutens/toxicity , Animals , Caco-2 Cells , Cell Differentiation , Cell Line , Duodenum/drug effects , Enterocytes/cytology , Gliadin/toxicity , Humans , In Vitro Techniques , Mice , Models, Biological , Triticum/chemistry
5.
Nutrients ; 12(2)2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32075195

ABSTRACT

Celiac disease (CD) is an autoimmune enteropathy caused by an intolerance to gluten proteins. It has been hypothesized that probiotic bacteria may exert beneficial effects by modulating inflammatory processes and by sustaining peptide hydrolysis at the intestinal level. This study aims at evaluating the capacity of a probiotic mixture (two different strains of lactobacilli and three of bifidobacteria) to hydrolyze gluten peptides following simulated gastrointestinal digestion of gliadin (PT-gliadin). The capacity of bacterial hydrolysates to counteract the toxic effects of gliadin-derived peptides in Caco-2 cells was also assessed. The protein and peptide mixtures, untreated or proteolyzed with the probiotic preparation, were analyzed before and after each proteolytic step with different techniques (SDS-PAGE, reverse phase HPLC, filtration on different molecular cut-off membranes). These experiments demonstrated that PT-gliadin can be further digested by bacteria into lower molecular weight peptides. PT-gliadin, untreated or digested with the probiotics, was then used to evaluate oxidative stress, IL-6 cytokine production and expression of tight junctions' proteins-such as occludin and zonulin-in Caco-2 cells. PT-gliadin induced IL-6 production and modulation and redistribution of zonulin and occludin, while digestion with the probiotic strains reversed these effects. Our data indicate that this probiotic mixture may exert a protective role in CD.


Subject(s)
Bifidobacterium , Gliadin/metabolism , Gliadin/toxicity , Lactobacillus , Probiotics/pharmacology , Protein Hydrolysates/pharmacology , Bifidobacterium/metabolism , Caco-2 Cells , Celiac Disease/prevention & control , Celiac Disease/therapy , Haptoglobins/metabolism , Humans , Hydrolysis , Interleukin-6/metabolism , Lactobacillus/metabolism , Molecular Weight , Occludin/metabolism , Oxidative Stress/drug effects , Probiotics/therapeutic use , Protein Precursors/metabolism , Proteolysis
6.
Gastroenterology ; 158(6): 1667-1681.e12, 2020 05.
Article in English | MEDLINE | ID: mdl-32032584

ABSTRACT

BACKGROUND & AIMS: Celiac disease could be treated, and potentially cured, by restoring T-cell tolerance to gliadin. We investigated the safety and efficacy of negatively charged 500-nm poly(lactide-co-glycolide) nanoparticles encapsulating gliadin protein (TIMP-GLIA) in 3 mouse models of celiac disease. Uptake of these nanoparticles by antigen-presenting cells was shown to induce immune tolerance in other animal models of autoimmune disease. METHODS: We performed studies with C57BL/6; RAG1-/- (C57BL/6); and HLA-DQ8, huCD4 transgenic Ab0 NOD mice. Mice were given 1 or 2 tail-vein injections of TIMP-GLIA or control nanoparticles. Some mice were given intradermal injections of gliadin in complete Freund's adjuvant (immunization) or of soluble gliadin or ovalbumin (ear challenge). RAG-/- mice were given intraperitoneal injections of CD4+CD62L-CD44hi T cells from gliadin-immunized C57BL/6 mice and were fed with an AIN-76A-based diet containing wheat gluten (oral challenge) or without gluten. Spleen or lymph node cells were analyzed in proliferation and cytokine secretion assays or by flow cytometry, RNA sequencing, or real-time quantitative polymerase chain reaction. Serum samples were analyzed by gliadin antibody enzyme-linked immunosorbent assay, and intestinal tissues were analyzed by histology. Human peripheral blood mononuclear cells, or immature dendritic cells derived from human peripheral blood mononuclear cells, were cultured in medium containing TIMP-GLIA, anti-CD3 antibody, or lipopolysaccharide (controls) and analyzed in proliferation and cytokine secretion assays or by flow cytometry. Whole blood or plasma from healthy volunteers was incubated with TIMP-GLIA, and hemolysis, platelet activation and aggregation, and complement activation or coagulation were analyzed. RESULTS: TIMP-GLIA did not increase markers of maturation on cultured human dendritic cells or induce activation of T cells from patients with active or treated celiac disease. In the delayed-type hypersensitivity (model 1), the HLA-DQ8 transgenic (model 2), and the gliadin memory T-cell enteropathy (model 3) models of celiac disease, intravenous injections of TIMP-GLIA significantly decreased gliadin-specific T-cell proliferation (in models 1 and 2), inflammatory cytokine secretion (in models 1, 2, and 3), circulating gliadin-specific IgG/IgG2c (in models 1 and 2), ear swelling (in model 1), gluten-dependent enteropathy (in model 3), and body weight loss (in model 3). In model 1, the effects were shown to be dose dependent. Splenocytes from HLA-DQ8 transgenic mice given TIMP-GLIA nanoparticles, but not control nanoparticles, had increased levels of FOXP3 and gene expression signatures associated with tolerance induction. CONCLUSIONS: In mice with gliadin sensitivity, injection of TIMP-GLIA nanoparticles induced unresponsiveness to gliadin and reduced markers of inflammation and enteropathy. This strategy might be developed for the treatment of celiac disease.


Subject(s)
Celiac Disease/drug therapy , Gliadin/administration & dosage , Immune Tolerance/drug effects , Nanoparticles/administration & dosage , Administration, Intravenous , Animals , CD4-Positive T-Lymphocytes , Celiac Disease/blood , Celiac Disease/immunology , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Gliadin/immunology , Gliadin/toxicity , Glutens/administration & dosage , Glutens/immunology , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , Humans , Intestinal Mucosa , Leukocytes, Mononuclear , Mice , Mice, Transgenic , Nanoparticles/chemistry , Nanoparticles/toxicity , Polyglactin 910/chemistry , Primary Cell Culture , Toxicity Tests, Acute
7.
Nutrients ; 11(11)2019 Oct 27.
Article in English | MEDLINE | ID: mdl-31717869

ABSTRACT

Gliadin is a major protein component of gluten and causes gluten toxicity through intestinal stress. We previously showed that gliadin intake induces oxidative stress in the intestine and reduces fertility in a Caenorhabditis elegans model. To elucidate the possible link between intestinal stress and reproduction, changes in the intestine and germ cells of C. elegans after gliadin intake were examined at the molecular level. Gliadin intake increased reactive oxygen species (ROS) production in the intestine, decreased intestinal F-actin levels, and increased germ cell apoptosis. These gliadin-triggered effects were suppressed by antioxidant treatment. These results suggest that ROS production in the intestine induced by gliadin intake causes disruption of intestinal integrity and increases germ cell apoptosis. Gliadin-induced germ cell apoptosis (GIGA) was suppressed by depletion of cep-1, ced-13, egl-1, or mpk-1. However, HUS-1 was not activated, suggesting that GIGA is activated through the mitogen-activated protein kinase (MAPK) pathway and is CEP-1-dependent but is a separate pathway from that controlling the DNA damage response. Taken together, our results suggest that gliadin causes intestinal barrier disruption through ROS production and interacts with the germ cells to reduce fertility through GIGA.


Subject(s)
Apoptosis/drug effects , Gliadin/toxicity , Intestinal Mucosa/drug effects , Animals , Caenorhabditis elegans , Cell Survival/drug effects , Cells, Cultured , Germ Cells/drug effects , Gliadin/chemistry , Intestinal Mucosa/metabolism , Mice , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
8.
Cells ; 8(4)2019 04 12.
Article in English | MEDLINE | ID: mdl-31013754

ABSTRACT

Celiac disease (CD) is a chronic systemic autoimmune disorder that is triggered by the ingestion of gliadin peptides, the alcohol-soluble fraction of wheat gluten. These peptides, which play a key role in the immune response that underlies CD, spontaneously form aggregates and exert a direct toxic action on cells due to the increase in the reactive oxygen species (ROS) levels. Furthermore, peptic-tryptic digested gliadin peptides (PT-gliadin) lead to an impairment in the autophagy pathway in an in vitro model based on Caco-2 cells. Considering these premises, in this study we have analyzed different mTOR-independent inducers, reporting that the disaccharide trehalose, a mTOR-independent autophagy activator, rescued the autophagy flux in Caco-2 cells treated with digested gliadin, as well as improved cell viability. Moreover, trehalose administration to Caco-2 cells in presence of digested gliadin reduced the intracellular levels of these toxic peptides. Altogether, these results showed the beneficial effects of trehalose in a CD in vitro model as well as underlining autophagy as a molecular pathway whose modulation might be promising in counteracting PT-gliadin cytotoxicity.


Subject(s)
Celiac Disease/metabolism , Trehalose/pharmacology , Autophagy/drug effects , Caco-2 Cells , Celiac Disease/immunology , Cell Survival/drug effects , Gliadin/adverse effects , Gliadin/chemistry , Gliadin/toxicity , Glutens , HT29 Cells , Humans , Models, Biological , Peptides , Reactive Oxygen Species , Trehalose/metabolism , Triticum/metabolism
9.
Aging (Albany NY) ; 11(7): 2003-2019, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30981209

ABSTRACT

In celiac disease (CD), an intolerance to dietary gluten/gliadin, antigenic gliadin peptides trigger an HLA-DQ2/DQ8-restricted adaptive Th1 immune response. Epithelial stress, induced by other non-antigenic gliadin peptides, is required for gliadin to become fully immunogenic. We found that cystic-fibrosis-transmembrane-conductance-regulator (CFTR) acts as membrane receptor for gliadin-derived peptide P31-43, as it binds to CFTR and impairs its channel function. P31-43-induced CFTR malfunction generates epithelial stress and intestinal inflammation. Maintaining CFTR in an active open conformation by the CFTR potentiators VX-770 (Ivacaftor) or Vrx-532, prevents P31-43 binding to CFTR and controls gliadin-induced manifestations. Here, we evaluated the possibility that the over-the-counter nutraceutical genistein, known to potentiate CFTR function, would allow to control gliadin-induced alterations. We demonstrated that pre-treatment with genistein prevented P31-43-induced CFTR malfunction and an epithelial stress response in Caco-2 cells. These effects were abrogated when the CFTR gene was knocked out by CRISP/Cas9 technology, indicating that genistein protects intestinal epithelial cells by potentiating CFTR function. Notably, genistein protected gliadin-sensitive mice from intestinal CFTR malfunction and gliadin-induced inflammation as it prevented gliadin-induced IFN-γ production by celiac peripheral-blood-mononuclear-cells (PBMC) cultured ex-vivo in the presence of P31-43-challenged Caco-2 cells. Our results indicate that natural compounds capable to increase CFTR channel gating might be used for the treatment of CD.


Subject(s)
Celiac Disease/prevention & control , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Genistein/pharmacology , Gliadin/toxicity , Peptide Fragments/toxicity , Animals , Caco-2 Cells , Celiac Disease/etiology , Celiac Disease/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dietary Supplements , Disease Models, Animal , Female , Gene Knockout Techniques , Gliadin/immunology , Humans , Interferon-gamma/biosynthesis , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Male , Mice , Mice, Inbred BALB C , Models, Biological , Peptide Fragments/immunology , Protein Binding
10.
Cell Death Dis ; 10(4): 258, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30874543

ABSTRACT

Under physiological conditions, a finely tuned system of cellular adaptation allows the intestinal mucosa to maintain the gut barrier function while avoiding excessive immune responses to non-self-antigens from dietary origin or from commensal microbes. This homeostatic function is compromised in cystic fibrosis (CF) due to loss-of-function mutations in the CF transmembrane conductance regulator (CFTR). Recently, we reported that mice bearing defective CFTR are abnormally susceptible to a celiac disease-like enteropathy, in thus far that oral challenge with the gluten derivative gliadin elicits an inflammatory response. However, the mechanisms through which CFTR malfunction drives such an exaggerated response to dietary protein remains elusive. Here we demonstrate that the proteostasis regulator/transglutaminase 2 (TGM2) inhibitor cysteamine restores reduced Beclin 1 (BECN1) protein levels in mice bearing cysteamine-rescuable F508del-CFTR mutant, either in homozygosis or in compound heterozygosis with a null allele, but not in knock-out CFTR mice. When cysteamine restored BECN1 expression, autophagy was increased and gliadin-induced inflammation was reduced. The beneficial effects of cysteamine on F508del-CFTR mice were lost when these mice were backcrossed into a Becn1 haploinsufficient/autophagy-deficient background. Conversely, the transfection-enforced expression of BECN1 in human intestinal epithelial Caco-2 cells mitigated the pro-inflammatory cellular stress response elicited by the gliadin-derived P31-43 peptide. In conclusion, our data provide the proof-of-concept that autophagy stimulation may mitigate the intestinal malfunction of CF patients.


Subject(s)
Autophagy/drug effects , Cysteamine/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/genetics , Gliadin/immunology , Peptide Fragments/immunology , Animals , Autophagy/genetics , Beclin-1/genetics , Beclin-1/metabolism , Caco-2 Cells , Cysteamine/therapeutic use , Cystic Fibrosis/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Gliadin/toxicity , Heterozygote , Homozygote , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/toxicity , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/antagonists & inhibitors , Transglutaminases/metabolism
11.
Int J Food Sci Nutr ; 69(8): 954-962, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29495907

ABSTRACT

Triticum monococcum L. is one of the oldest ancestors of wheat. There is some evidence that einkorn encloses forms of gliadin-deriving peptides which may potentially exert a reduced toxicity to consumers with gluten-related disorders. Accordingly, ID331 and Monlis lines were comparatively investigated in this study. The biological effects of gastro-resistant peptides deriving from an in vitro simulated digestion were evaluated on 21 d differentiated Caco-2 cells. Triticum aestivum digested gliadin was included as the positive control. ID331 neither enhanced cell permeability nor induced zonulin release in Caco-2 monolayers. Monlis exerted a detectable toxicity as confirmed by the reorganisation of enterocyte cytoskeleton, in addition to changes both in monolayers permeability and apical release of zonulin. Differences in patterns of gastro-resistant prolamins may account for the differences. Outcomes support the use of ID331 as a prospective candidate for the development of innovative approaches to reduce wheat flour toxicity.


Subject(s)
Gliadin/toxicity , Triticum/chemistry , Triticum/classification , Actins/metabolism , Caco-2 Cells , Celiac Disease/prevention & control , Cell Survival/drug effects , Cholera Toxin/metabolism , Flour , Glutens/immunology , Haptoglobins , Humans , Permeability , Protein Precursors , Triticum/toxicity
12.
Cell Biol Int ; 42(1): 112-120, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28914468

ABSTRACT

Alpha-gliadin peptide 31-43 is considered to be the main peptide responsible for the innate immune response in celiac disease patients. Recent evidence indicates that peptide 31-43 rapidly enters cells and interacts with the early endocytic vesicular compartment. However, the mechanism of its uptake is not completely understood. Our aim is to characterize, isolate and identify possible cell surface proteins involved in peptide 31-43 internalization by Caco-2 cells. In this study, we used a chemical cross-linker to block peptide 31-43 on cell surface proteins, and pulled-down peptide-proteins complexes using antibodies raised against peptide 31-43. Through this experimental approach, we did not observe any specific complex between cell proteins and peptide 31-43 in Coomassie-stained denaturating gels or by Western blotting. We also found that type 2 transglutaminase was not necessary for peptide 31-43 internalization, even though it had a regulatory role in the process. Finally, we demonstrated that peptide 31-43 did not behave as a classical ligand, indeed the labeled peptide did not displace the unlabeled peptide in a competitive binding assay. On the basis of these findings and of previous evidence demonstrating that peptide 31-43 is able to interact with a membrane-like environment in vitro, we conclude that membrane composition and organization, rather than a specific receptor protein, may have a major role in peptide 31-43 internalization by cells.


Subject(s)
Endocytosis/physiology , Gliadin/metabolism , Antibodies/immunology , Caco-2 Cells/metabolism , Celiac Disease/immunology , Celiac Disease/physiopathology , Cell Count , GTP-Binding Proteins , Gliadin/toxicity , HEK293 Cells/metabolism , Humans , Immunity, Innate/immunology , Immunity, Innate/physiology , Intestinal Mucosa/metabolism , Peptide Fragments/metabolism , Peptides/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Receptors, Cell Surface/physiology , Transglutaminases
13.
Am J Physiol Gastrointest Liver Physiol ; 311(1): G40-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27151946

ABSTRACT

Celiac disease (CD) is an immune-mediated enteropathy triggered by gluten in genetically susceptible individuals. Innate immunity contributes to the pathogenesis of CD, but the mechanisms remain poorly understood. Although previous in vitro work suggests that gliadin peptide p31-43 acts as an innate immune trigger, the underlying pathways are unclear and have not been explored in vivo. Here we show that intraluminal delivery of p31-43 induces morphological changes in the small intestinal mucosa of normal mice consistent with those seen in CD, including increased cell death and expression of inflammatory mediators. The effects of p31-43 were dependent on MyD88 and type I IFNs, but not Toll-like receptor 4 (TLR4), and were enhanced by coadministration of the TLR3 agonist polyinosinic:polycytidylic acid. Together, these results indicate that gliadin peptide p31-43 activates the innate immune pathways in vivo, such as IFN-dependent inflammation, relevant to CD. Our findings also suggest a common mechanism for the potential interaction between dietary gluten and viral infections in the pathogenesis of CD.


Subject(s)
Celiac Disease/immunology , Gliadin/toxicity , Immunity, Innate/drug effects , Immunity, Mucosal/drug effects , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Peptide Fragments/toxicity , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Celiac Disease/metabolism , Celiac Disease/pathology , Gene Expression Regulation , Genotype , Gliadin/administration & dosage , Inflammation Mediators/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Peptide Fragments/administration & dosage , Phenotype , Poly I-C/pharmacology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Signal Transduction/drug effects , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism
14.
Pediatr Res ; 78(4): 358-64, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26086640

ABSTRACT

BACKGROUND: In Celiac disease (CD), cytoskeletal integrity of intestinal cells is disrupted by gliadin exposure. This study investigates the role of heat shock protein (Hsp)70 during cytoskeletal recovery in CD by assessing its induction and effects on junctional proteins. METHODS: Using an in-vitro model of CD, cytoskeletal injury and recovery was assessed in gliadin-exposed Caco-2 cells by measuring cellular distribution of ezrin, E-cadherin, and Hsp70 by differential centrifugation. Effects of Hsp70 were tested by an in-vitro repair assay, based on the incubation of injured or recovered cytoskeletal cellular fractions in noncytoskeletal supernatants containing low or high levels of Hsp70, or by transient transfection of Caco-2 cells with Hsp70. RESULTS: Cytoskeletal disruption of ezrin and E-cadherin was demonstrated in gliadin-exposed Caco-2 cells by their significant shift from the cytoskeletal pellet into the noncytoskeletal supernatant fraction. Recovery from gliadin exposure was associated with induction and cytoskeletal redistribution of Hsp70. The in-vitro repair assay delineated direct evidence for HSP-mediated repair by stabilization of junctional proteins by Hsp70. Overexpression of Hsp70 resulted in significantly increased cytoskeletal integrity. CONCLUSION: Our results establish an essential role of HSP-mediated cytoskeletal repair in Caco-2 cells during recovery from in-vitro gliadin exposure.


Subject(s)
Celiac Disease/metabolism , Epithelial Cells/drug effects , Gliadin/toxicity , HSP70 Heat-Shock Proteins/metabolism , Intestinal Mucosa/drug effects , Antigens, CD , Caco-2 Cells , Cadherins/metabolism , Celiac Disease/genetics , Celiac Disease/pathology , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , HSP70 Heat-Shock Proteins/genetics , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Protein Transport , Signal Transduction/drug effects , Time Factors , Transfection , Up-Regulation
15.
Clin Exp Med ; 15(4): 447-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25447031

ABSTRACT

The downstream cascade of the inflammatory response to gliadin in celiac intestinal mucosa encompasses the early activation of the innate immunity that triggers the adaptive response. Therefore, the in vitro study of the pathogenic mechanism of celiac disease (CD) on enterocytes alone or mucosal T lymphocytes alone does not fully consider all the aspects of gliadin-dependent inflammation. Although the in vitro culture of specimens of intestinal mucosa obtained from celiac patients is the gold standard for the study of CD, this technique presents several technical challenges and the bioptic specimens are not easily available. So, in this paper, we described the gliadin-dependent cytokine production in a bidimensional cellular system, which is able to mimic both the innate and the adaptive steps of the mucosal immune response of CD. In the upper compartment, the intestinal epithelial cells are grown on a filter, and in the lower compartment, the mononuclear cells isolated from peripheral blood of celiac patients are cultured. Cells were apically exposed to the toxic gliadin peptide p31-43 for 3 h and then with the immunodominant gliadin fragment pα-9 for 21 h. The incubation with gliadin peptides resulted in increased levels of IL-15, INF-γ, IL-6, tumor necrosis factor (TNF)-α, IL-1ß, and CCL 2, 3 and 4 in the basal supernatants, with respect to cells exposed to medium alone. The p31-43-driven epithelial priming of mucosal response consists of transglutaminase (TG2)-mediated deamidation of the immunostimulatory gliadin peptides, as demonstrated by the inhibition of pα-9 activity, when the system is exposed to blocking anti-TG2 antibody.


Subject(s)
Celiac Disease/pathology , Cytokines/metabolism , Gliadin/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Adaptive Immunity , Adolescent , Cell Culture Techniques , Cells, Cultured , Child , Cytokines/biosynthesis , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Gliadin/toxicity , Humans , Immunity, Innate , Male , Models, Biological , Monocytes/physiology
16.
PLoS One ; 9(11): e113932, 2014.
Article in English | MEDLINE | ID: mdl-25415429

ABSTRACT

SCOPE: Celiac disease is an autoimmune disorder caused by failure of oral tolerance against gluten in genetically predisposed individuals. The epithelial translocation of gluten-derived gliadin peptides is an important pathogenetic step; the underlying mechanisms, however, are poorly understood. Thus, we investigated the degradation and epithelial translocation of two different gliadin peptides, the toxic P31-43 and the immunogenic P56-68. As the size, and hence, the molecular weight of peptides might have an effect on the transport efficiency we chose two peptides of the same, rather short chain length. METHODS AND RESULTS: Fluorescence labeled P31-43 and P56-68 were synthesized and studied in a transwell system with human enterocytes. Fluorometric measurements were done to reveal antigen translocation and flow cytometry as well as confocal microscopy were used to investigate cellular uptake of peptides. Structural changes of these peptides were analysed by MALDI-TOF-MS. According to fluorescence intensities, significantly more P31-43 compared to P56-68 was transported through the enterocyte layer after 24 h incubation. In contrast to previous reports, however, mass spectrometric data do not only show a time-dependent cleavage of the immunogenic P56-68, but we observed for the first time the degradation of the toxic peptide P31-43 at the apical side of epithelial cells. CONCLUSION: Considering the degradation of gliadin peptides by enterocytes, measurement of fluorescence signals do not completely represent translocated intact gliadin peptides. From our experiments it is obvious that even short peptides can be digested prior to the translocation across the epithelial barrier. Thus, the chain length and the sensibility to degradations of gliadin peptides as well as the integrity of the epithelial barrier seem to be critical for the uptake of gliadin peptides and the subsequent inflammatory immune response.


Subject(s)
Enterocytes/metabolism , Gliadin/pharmacokinetics , Gliadin/toxicity , Peptide Fragments/pharmacokinetics , Peptide Fragments/toxicity , Caco-2 Cells , Celiac Disease/immunology , Celiac Disease/metabolism , Celiac Disease/pathology , Enterocytes/immunology , Enterocytes/pathology , Humans , Protein Transport/drug effects
17.
BMC Microbiol ; 14: 19, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24483336

ABSTRACT

BACKGROUND: Celiac disease is characterized by enhanced intestinal paracellular permeability due to alterations of function and expression of tight junction (TJ) proteins including ZO-1, Claudin-1 and Occludin. Polyamines are pivotal in the control of intestinal barrier function and are also involved in the regulation of intercellular junction proteins. Different probiotic strains may inhibit gliadin-induced toxic effects and the Lactobacillus rhamnosus GG (L.GG) is effective in the prevention and treatment of gastrointestinal diseases. Aims of the study were to establish in epithelial Caco-2 cells whether i) gliadin affects paracellular permeability and polyamine profile; ii) co-administration of viable L.GG, heat-killed L.GG (L.GG-HK) or its conditioned medium (L.GG-CM) preserves the intestinal epithelial barrier integrity. Additionally, the effects of L.GG on TJ protein expression were tested in presence or absence of polyamines. RESULTS: Administration of gliadin (1 mg/ml) to Caco-2 cells for 6 h caused a significant alteration of paracellular permeability as demonstrated by the rapid decrease in transepithelial resistance with a concomitant zonulin release. These events were followed by a significant increase in lactulose paracellular transport and a slight lowering in ZO-1 and Occludin expression without affecting Claudin-1. Besides, the single and total polyamine content increased significantly. The co-administration of viable L.GG (10(8) CFU/ml), L.GG-HK and L.GG-CM with gliadin significantly restored barrier function as demonstrated by transepithelial resistance, lactulose flux and zonulin release. Viable L.GG and L.GG-HK, but not L.GG-CM, led to a significant reduction in the single and total polyamine levels. Additionally, only the co-administration of viable L.GG with gliadin significantly increased ZO-1, Claudin-1 and Occludin gene expression compared to control cells. When Caco-2 cells treated with viable L.GG and gliadin were deprived in the polyamine content by α-Difluoromethylornithine, the expression of TJ protein mRNAs was not significantly different from that in controls or cells treated with gliadin alone. CONCLUSIONS: Gliadin modifies the intestinal paracellular permeability and significantly increases the polyamine content in Caco-2 cells. Concomitant administration of L.GG is able to counteract these effects. Interestingly, the presence of cellular polyamines is necessary for this probiotic to exert its capability in restoring paracellular permeability by affecting the expression of different TJ proteins.


Subject(s)
Epithelial Cells/drug effects , Gliadin/toxicity , Lacticaseibacillus rhamnosus/physiology , Polyamines/metabolism , Probiotics , Tight Junctions/drug effects , Caco-2 Cells , Epithelial Cells/metabolism , Epithelial Cells/physiology , Humans , Permeability/drug effects
18.
PLoS One ; 8(11): e79763, 2013.
Article in English | MEDLINE | ID: mdl-24278174

ABSTRACT

Celiac disease (CD) is a frequent inflammatory intestinal disease, with a genetic background, caused by gliadin-containing food. Undigested gliadin peptides P31-43 and P57-68 induce innate and adaptive T cell-mediated immune responses, respectively. Alterations in the cell shape and actin cytoskeleton are present in celiac enterocytes, and gliadin peptides induce actin rearrangements in both the CD mucosa and cell lines. Cell shape is maintained by the actin cytoskeleton and focal adhesions, sites of membrane attachment to the extracellular matrix. The locus of the human Lipoma Preferred Partner (LPP) gene was identified as strongly associated with CD using genome-wide association studies (GWAS). The LPP protein plays an important role in focal adhesion architecture and acts as a transcription factor in the nucleus. In this study, we examined the hypothesis that a constitutive alteration of the cell shape and the cytoskeleton, involving LPP, occurs in a cell compartment far from the main inflammation site in CD fibroblasts from skin explants. We analyzed the cell shape, actin organization, focal adhesion number, focal adhesion proteins, LPP sub-cellular distribution and adhesion to fibronectin of fibroblasts obtained from CD patients on a Gluten-Free Diet (GFD) and controls, without and with treatment with A-gliadin peptide P31-43. We observed a "CD cellular phenotype" in these fibroblasts, characterized by an altered cell shape and actin organization, increased number of focal adhesions, and altered intracellular LPP protein distribution. The treatment of controls fibroblasts with gliadin peptide P31-43 mimics the CD cellular phenotype regarding the cell shape, adhesion capacity, focal adhesion number and LPP sub-cellular distribution, suggesting a close association between these alterations and CD pathogenesis.


Subject(s)
Celiac Disease/metabolism , Cytoskeletal Proteins/metabolism , Gliadin/toxicity , LIM Domain Proteins/metabolism , Peptide Fragments/toxicity , Cells, Cultured , Diet, Gluten-Free/adverse effects , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Genome-Wide Association Study , Humans , Male
19.
J Proteomics ; 77: 310-20, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23023000

ABSTRACT

Celiac disease is an immune-mediated disorder triggered by gluten proteins of wheat (gliadins) and other cereals. Gliadin-mediated effects on weanling animals, sensitized or not with interferon (IFN)-γ, were investigated. Also, the influence of the co-administration of Bifidobacterium longum CECT 7347 was studied together with changes in the proteome of jejunal sections, using 2DE and MALDITOF-TOF peptide fingerprinting. Findings were compared to results for control animal groups. In the principal component analysis (PCA) of proteome pattern, two components were extracted accounting for 79.8% of variability in the expression of the identified proteins. PCA analysis clearly discriminated between the proteome of animals fed gliadins alone and those fed gliadins and B. longum simultaneously. However, the proteome patterns from animals sensitized with IFN-γ and fed gliadins together with B. longum, or alone, could not be discriminated. Gliadin feeding caused inflammatory effects as well as changes in proteins involved in intracellular ionic homeostasis, lipid turnover, cell motility and redox regulation in intestinal sections. After feeding gliadins to animals sensitized with IFN-γ, changes were also detected in proteins involved in recruitment and function of inmunocompetent cells, trophic effect on the intestine and organization of myofibers reflecting the more marked gliadin-mediated injury in jejunal sections. The administration of the bacterial strain to rats fed gliadins seemed to ameliorate the inflammation caused by gliadin feeding alone, although, in sensitized animals the co-administration of B. longum had less marked effects, which was probably due to the more extensive intestinal mucosal damage. The proteome patterns in animals administered B. longum alone did not reveal any changes reflecting impairment of jejunal functions.


Subject(s)
Bifidobacterium , Celiac Disease/metabolism , Gliadin/toxicity , Intestinal Mucosa/metabolism , Jejunum/metabolism , Probiotics/pharmacology , Proteome/metabolism , Animals , Antiviral Agents/adverse effects , Antiviral Agents/pharmacology , Celiac Disease/chemically induced , Celiac Disease/microbiology , Celiac Disease/pathology , Disease Models, Animal , Female , Humans , Infant , Interferon-gamma/adverse effects , Interferon-gamma/pharmacology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Jejunum/microbiology , Jejunum/pathology , Male , Rats , Rats, Wistar
20.
Gastroenterology ; 142(2): 316-25.e1-12, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22079593

ABSTRACT

BACKGROUND & AIMS: Copolymers of hydroxyethyl methacrylate and styrene sulfonate complex with isolated gliadin (the toxic fraction of gluten) and prevent damage to the intestinal barrier in HLA-HCD4/DQ8 mice. We studied the activity toward gluten and hordein digestion and biologic effects of poly(hydroxyethyl methacrylate-co-styrene sulfonate (P(HEMA-co-SS)). We also investigated the effect of gliadin complex formation in intestinal biopsy specimens from patients with celiac disease. METHODS: We studied the ability of P(HEMA-co-SS) to reduce digestion of wheat gluten and barley hordein into immunotoxic peptides using liquid chromatography-mass spectrometry. The biodistribution and pharmacokinetic profile of orally administered P(HEMA-co-SS) was established in rodents using tritium-labeled polymer. We assessed the capacity of P(HEMA-co-SS) to prevent the immunologic and intestinal effects induced by a gluten-food mixture in gluten-sensitized HLA-HCD4/DQ8 mice after short-term and long-term administration. We measured the effects of gliadin complex formation on cytokine release ex vivo using intestinal biopsy specimens from patients with celiac disease. RESULTS: P(HEMA-co-SS) reduced digestion of wheat gluten and barley hordein in vitro, thereby decreasing formation of toxic peptides associated with celiac disease. After oral administration to rodents, P(HEMA-co-SS) was predominantly excreted in feces, even in the presence of low-grade mucosal inflammation and increased intestinal permeability. In gluten-sensitized mice, P(HEMA-co-SS) reduced paracellular permeability, normalized anti-gliadin immunoglobulin A in intestinal washes, and modulated the systemic immune response to gluten in a food mixture. Furthermore, incubation of P(HEMA-co-SS) with mucosal biopsy specimens from patients with celiac disease showed that secretion of tumor necrosis factor-α was reduced in the presence of partially digested gliadin. CONCLUSIONS: The copolymer P(HEMA-co-SS) reduced digestion of wheat gluten and barley hordein and attenuated the immune response to gluten in a food mixture in rodents. It might be developed to prevent or reduce gluten-induced disorders in humans.


Subject(s)
Celiac Disease/metabolism , Digestion/drug effects , Glutens/metabolism , Intestinal Mucosa/drug effects , Polyhydroxyethyl Methacrylate/analogs & derivatives , Polystyrenes/pharmacology , Styrenes/pharmacology , Animals , Celiac Disease/drug therapy , Celiac Disease/immunology , Chromatography, Liquid , Female , Gliadin/metabolism , Gliadin/toxicity , Glutens/toxicity , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Jejunum/drug effects , Jejunum/immunology , Jejunum/pathology , Male , Mass Spectrometry , Mice , Permeability/drug effects , Polyhydroxyethyl Methacrylate/pharmacokinetics , Polyhydroxyethyl Methacrylate/pharmacology , Polyhydroxyethyl Methacrylate/therapeutic use , Polystyrenes/pharmacokinetics , Polystyrenes/therapeutic use , Protein Binding , Random Allocation , Rats , Styrenes/pharmacokinetics , Styrenes/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...