Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Int J Mol Sci ; 23(4)2022 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-35216089

RESUMEN

Celiac disease (CD) is a chronic inflammatory disease caused by a genetic predisposition to an abnormal T cell-mediated immune response to the gluten in the diet. Different environmental proinflammatory factors can influence and amplify the T cell-mediated response to gluten. The aim of this manuscript was to study the role of enterocytes in CD intestinal inflammation and their response to different proinflammatory factors, such as gliadin and viruses. Intestinal biopsies from CD patients on a gluten-containing (GCD-CD) or a gluten-free diet (GFD-CD) as well as biopsies from potential CD patients (Pot-CD) before the onset of intestinal lesions and controls (CTR) were used to investigate IL-1ß and IL-6 mRNA levels in situ. Organoids from CD patients were used to test the levels of NF-κB, ERK, IL-6, and IL-1ß by Western blot (WB), ELISA, and quantitative PCR. The Toll-like receptor ligand loxoribine (Lox) and gliadin peptide P31-43 were used as proinflammatory stimuli. In CD biopsies inflammation markers IL-1ß and IL-6 were increased in the enterocytes, and also in Pot-CD before the onset of the intestinal lesion and in GFD-CD. The inflammatory markers pNF-κB, pERK, IL-1ß, and IL-6 were increased and persistent in CD organoids; these organoids were more sensitive to P31-43 and Lox stimuli compared with CTR organoids. Taken together, these observations point to constitutive inflammation in CD enterocytes, which are more sensitive to inflammatory stimuli such as food components and viruses.


Asunto(s)
Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/patología , Enterocitos/metabolismo , Enterocitos/patología , Inflamación/metabolismo , Inflamación/patología , Adolescente , Biomarcadores/metabolismo , Niño , Preescolar , Dieta Sin Gluten , Femenino , Glútenes/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Transducción de Señal/fisiología
2.
Int J Mol Sci ; 22(5)2021 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-33800150

RESUMEN

Celiac disease (CD) is a frequent intestinal inflammatory disease occurring in genetically susceptible individuals upon gluten ingestion. Recent studies point to a role in CD for genes involved in cell shape, adhesion and actin rearrangements, including a Rho family regulator, Rho GTPase-activating protein 31 (ARHGAP31). In this study, we investigated the morphology and actin cytoskeletons of peripheral monocyte-derived dendritic cells (DCs) from children with CD and controls when in contact with a physiological substrate, fibronectin. DCs were generated from peripheral blood monocytes of pediatric CD patients and controls. After adhesion on fibronectin, DCs showed a higher number of protrusions and a more elongated shape in CD patients compared with controls, as assessed by immunofluorescence actin staining, transmitted light staining and video time-lapse microscopy. These alterations did not depend on active intestinal inflammation associated with gluten consumption and were specific to CD, since they were not found in subjects affected by other intestinal inflammatory conditions. The elongated morphology was not a result of differences in DC activation or maturation status, and did not depend on the human leukocyte antigen (HLA)-DQ2 haplotype. Notably, we found that ARH-GAP31 mRNA levels were decreased while RhoA-GTP activity was increased in CD DCs, pointing to an impairment of the Rho pathway in CD cells. Accordingly, Rho inhibition was able to prevent the cytoskeleton rearrangements leading to the elongated morphology of celiac DCs upon adhesion on fibronectin, confirming the role of this pathway in the observed phenotype. In conclusion, adhesion on fibronectin discriminated CD from the controls' DCs, revealing a gluten-independent CD-specific cellular phenotype related to DC shape and regulated by RhoA activity.


Asunto(s)
Actinas/metabolismo , Enfermedad Celíaca/metabolismo , Forma de la Célula , Células Dendríticas/inmunología , Monocitos/metabolismo , Enfermedad Celíaca/patología , Adhesión Celular , Niño , Preescolar , Células Dendríticas/patología , Femenino , Fibronectinas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Antígenos HLA-DQ/metabolismo , Humanos , Masculino , Monocitos/patología , Fosfoproteínas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
3.
Int J Food Sci Nutr ; 70(8): 950-958, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30969137

RESUMEN

Coeliac disease is an increasingly recognised pathology, induced by the ingestion of gluten in genetically predisposed patients. Undigested gliadin peptide can induce adaptive and innate immune response that unleash the typical intestinal mucosal alterations. A growing attention is paid to alternative therapeutic approaches to the gluten-free diet: one of these approaches is the use of probiotics and/or postbiotics. We performed lactic fermentation of rice flour with and without pH control, using Lactobacillus paracasei CBA L74 as fermenting strain. We evaluated bacterial growth, lactic acid production during fermentation and gliadin peptide P31-43 entrance in CaCo-2 cells with and without pH control. When pH control was applied no differences were observed in terms of bacterial growth; on the contrary, lactic acid production was greater, as expected. Both samples could inhibit the P31-43 entrance in CaCo-2 cells but the effect was significantly greater for samples obtained when the pH control was applied.


Asunto(s)
Células Epiteliales/metabolismo , Fermentación , Gliadina/metabolismo , Concentración de Iones de Hidrógeno , Oryza/microbiología , Fragmentos de Péptidos/metabolismo , Células CACO-2 , Enfermedad Celíaca/tratamiento farmacológico , Enfermedad Celíaca/prevención & control , Dieta Sin Gluten , Hipersensibilidad a los Alimentos/prevención & control , Alimentos Funcionales , Gliadina/antagonistas & inhibidores , Glútenes , Humanos , Ácido Láctico/metabolismo , Lacticaseibacillus paracasei/metabolismo , Oryza/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores
4.
Am J Gastroenterol ; 111(1): 134-44, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26753888

RESUMEN

OBJECTIVES: Potential celiac disease (CD) patients are at an increased risk to developing CD as indicated by positive CD-associated serology. We investigated in duodenal mucosa of such patients the presence of both IL-21 and IL-17A and the role of gliadin peptides and IL-15 in their expression. METHODS: Duodenal biopsies from 76 active CD, 90 potential CD, and 58 control patients were analyzed for IL-21 and/or IL-17A production by quantitative real-time PCR, immunohistochemistry, flow cytometry, and ELISA. The presence of IL-21 receptor was investigated by western blot. Potential CD duodenal fragments were cultured with gliadin peptides (PTG) and/or IL-15 and the expression/production of IL-21 and IL-17A assessed by quantitative real-time PCR and by immunohistochemistry. RESULTS: In potential CD, IL-21 was lower than in active CD, in terms of RNA expression (P<0.01), density of lamina propria (LP) IL-21(+) cells (P<0.05), and protein secretion (P<0.05). Also, IL-21R was weakly detectable in potential CD. Several LP cell types produced IL-21 in CD. In potential CD, CD4(+)IL-21(+) cells increased after PMA-ionomycin stimulation and co-produced IFN-γ but not IL-17A. After 24 hours of culture stimulation with PTG, IL-21-producing cells increased but not the ones producing IL-17A. This increase was further enhanced by the addition of IL-15 to culture medium. CONCLUSIONS: In potential CD, IL-21 is less expressed than in active CD; however, IL-21-producing cells are present and prone to respond after specific stimuli. This suggests a key role of IL-21 in the progression of mucosal damage in CD.


Asunto(s)
Enfermedad Celíaca/metabolismo , Interleucina-17/biosíntesis , Interleucinas/biosíntesis , Mucosa Intestinal/metabolismo , Células Cultivadas , Niño , Preescolar , Duodeno/metabolismo , Femenino , Humanos , Masculino
5.
Int J Food Sci Nutr ; 65(8): 953-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25030417

RESUMEN

Several recent reports describe a role of probiotics as a therapeutic approach for celiac disease (CD). Two undigested A-gliadin peptides, P31-43 and P57-68, are central to CD pathogenesis, inducing an innate and an adaptive immune response, respectively. They enter enterocytes and localize to vesicular compartment to induce their toxic/immunogenics effects. In this article, we tested the effect of probiotic Lactobacillus paracasei (LP) CBA L74 (International Depository Accession Number LMG P-24778), its supernatant and LP-fermented cereals on gliadin peptides, P31-43 and P57-68, entrance in Caco-2 cells. Both LP CBA L74 and its supernatant inhibit P31-43 (intensity of fluorescence; FI: 75%) and P57-68 (FI: 50%) entrance in Caco2 cells, indicating that this biological effect is due to some product included in LP CBA L74 supernatant. This effect was present also after fermentation of cereals. This study describes a novel effect of probiotics in the prevention of undigested gliadin peptides toxic effects.


Asunto(s)
Productos Biológicos/farmacología , Enfermedad Celíaca/metabolismo , Gliadina/metabolismo , Mucosa Intestinal/metabolismo , Lactobacillus , Péptidos/metabolismo , Probióticos , Productos Biológicos/uso terapéutico , Células CACO-2 , Enfermedad Celíaca/tratamiento farmacológico , Células Cultivadas , Colon/efectos de los fármacos , Colon/metabolismo , Grano Comestible/microbiología , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Fermentación , Humanos , Mucosa Intestinal/efectos de los fármacos , Probióticos/uso terapéutico
6.
Am J Gastroenterol ; 108(11): 1775-84, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24060758

RESUMEN

OBJECTIVES: Potential celiac disease (CD) relates to subjects with a normal small intestinal mucosa who are at increased risk of developing CD as indicated by positive CD-associated serology. The objective of this study was to investigate in the small intestinal mucosa of such patients the state of immunological activation with special emphasis on immunoregulatory circuits. METHODS: Duodenal biopsies from active CD (n=48), potential CD (n=58), and control patients (n=45) were studied. RNA expression for interferon γ (IFNγ) and interleukin-10 (IL-10) were quantified by real-time quantitative PCR. The percentage of CD4+CD25+Foxp3+ T regulatory cells (Foxp3+Tregs) was determinated by flow cytometry and the number of Foxp3+ and IL-15+ cells by immunohistochemistry. Furthermore, we analyzed the suppressive function of CD4+CD25+ T cells, isolated from potential CD biopsy samples, as well as the effect of IL-15, on autologous peripheral blood responder CD4+CD25- T cells. RESULTS: In potential CD patients with Marsh 1 lesion, IFNγ-RNA expression was significantly less than in active, but enhanced if compared with potential CD patients with Marsh 0 lesion and with controls (P<0.001). The number of IL-15+ cells in subjects with potential CD was increased in comparison with controls (P<0.05), but lower than active CD (P<0.01). IL-10-RNA expression was upregulated in Marsh 0 potential CD patients if compared with those with Marsh 1 lesion (P<0.01) and controls (P<0.001), whereas there were no differences with active CD. The ratio IL-10/IFNγ reached the highest value in Marsh 0 potential CD compared with the other groups (P<0.05). The percentage of Foxp3+Tregs was also higher in potential CD compared with controls (P<0.05), although it was lower than in active CD (P<0.01). In co-culture assay, intestinal CD4+CD25+ T cells from potential CD patients exerted suppressive effects on T responder cells, and their activity was not impaired by IL-15. CONCLUSIONS: Potential CD patients show a low grade of inflammation that likely could be due to active regulatory mechanisms preventing the progression toward a mucosal damage.


Asunto(s)
Enfermedad Celíaca/inmunología , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/patología , Niño , Preescolar , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-15/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
7.
Cells ; 12(1)2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36611909

RESUMEN

BACKGROUND & AIMS: Celiac disease (CeD) is an immune-mediated enteropathy triggered in genetically susceptible (HLA-DQ2/8) individuals by a group of wheat proteins and related prolamins from cereals. The celiac intestine is characterized by an inversion of the differentiation/proliferation program of the enterocytes, with an increase in the proliferative compartment and crypt hyperplasia, which are the mechanisms that regulate the increased proliferation in CeD that arenot completely understood.The aim of this study is to understand the role of Protein Tyrosine Phosphatase Receptor Type K (PTPRK), a nodal phosphatase that regulates EGFR activation in the proliferation of the enterocytes from CeD biopsies and organoids. METHODS: The levels of PTPRK were evaluated by RT PCR, western blot (WB) and immunofluorescence techniques in intestinal biopsies and organoids from CeD patients and controls. Additionally, pEGFR and pERK were evaluated by WB and proliferation by BrdU incorporation. PTPRK si-RNA was silenced in CTR organoids and was overexpressed in CeD organoids. RESULTS: PTPRK was reduced in Gluten Containing Diet-Celiac Disease (GCD-CeD) and Potential-Celiac Disease(Pot-CeD) biopsies (p < 0.01-p < 0.05) whereas pEGFR (p < 0.01 p < 0.01), pERK (p < 0.01 p < 0.01) and proliferation were increased. (p < 0.05 p < 0.05) respect to the controls.The CeD organoids reproduced these same alterations. Silencing of PTPRK in CTR organoids increased pEGFR, pERK and proliferation. The overexpression of PTPRK in CeD organoids reduced pEGFR, pERK and proliferation. CONCLUSIONS: modulation of PTPRK levels can reduce or increase pEGFR, pERK and proliferation in CeD or CTR organoids, respectively. The CeD organoids can be a good model to study the mechanisms of the disease.


Asunto(s)
Enfermedad Celíaca , Humanos , Enfermedad Celíaca/genética , Enfermedad Celíaca/metabolismo , Receptores ErbB/metabolismo , Enterocitos/metabolismo , Biopsia , Predisposición Genética a la Enfermedad , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo
8.
Am J Gastroenterol ; 106(7): 1308-17, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21468011

RESUMEN

OBJECTIVES: Celiac disease (CD) is a condition in which the regulation of the mucosal immune response to dietary gliadin might be altered. The transcription factor forkhead box P3 (Foxp3) has been identified as a marker of a subset of regulatory T cells (Treg). In this study, we have investigated the presence and the suppressive function of Treg cells in the celiac small intestinal mucosa, their correlation with the disease state, and the inducibility by gliadin in an organ culture system; moreover, we tried to define whether interleukin 15 (IL-15), overexpressed in CD, could influence the regulatory activity of such cells. METHODS: The expression of Foxp3, CD3, CD4, and CD8 were analyzed by immunohistochemistry and flow cytometry in duodenal biopsies taken from patients with untreated CD, treated CD, and from non-CD controls, as well as in vitro cultured biopsy samples from treated CD patients, upon challenge with gliadin. Furthermore, we analyzed the suppressive function of CD4+CD25+ T cells, isolated from untreated CD biopsy samples, on autologous responder CD4+CD25- T cells, in the presence of a polyclonal stimulus, with or without IL-15. RESULTS: Higher density of CD4+CD25+Foxp3+ T cells was seen in duodenal biopsy samples from active CD patients in comparison with treated CD and non-CD controls. In coculture, CD4+CD25+ T cells were functionally suppressive, but their activity was impaired by IL-15. Cells from CD subjects showed increased sensitivity to the IL-15 action, likely due to enhanced expression of IL-15 receptor. Finally, we demonstrated an expansion of Foxp3 in treated CD mucosa following in vitro challenge with gliadin. CONCLUSIONS: These data suggest that CD4+CD25+Foxp3+ T cells are induced in situ by gliadin. However, their suppressor capacity might be impaired in vivo by IL-15; this phenomenon contributes to maintain and expand the local inflammatory response in CD.


Asunto(s)
Enfermedad Celíaca/metabolismo , Factores de Transcripción Forkhead/metabolismo , Gliadina/farmacología , Interleucina-15/farmacología , Mucosa Intestinal/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Adolescente , Adulto , Complejo CD3/metabolismo , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Enfermedad Celíaca/tratamiento farmacológico , Células Cultivadas , Duodeno/metabolismo , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Masculino , Persona de Mediana Edad , Linfocitos T Reguladores/inmunología , Adulto Joven
10.
Commun Biol ; 2: 190, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31123714

RESUMEN

Celiac Disease (CD) is an autoimmune disease characterized by inflammation of the intestinal mucosa due to an immune response to wheat gliadins. Some gliadin peptides (e.g., A-gliadin P57-68) induce an adaptive Th1 pro-inflammatory response. Other gliadin peptides (e.g., A-gliadin P31-43) induce a stress/innate immune response involving interleukin 15 (IL15) and interferon α (IFN-α). In the present study, we describe a stressed/inflamed celiac cellular phenotype in enterocytes and fibroblasts probably due to an alteration in the early-recycling endosomal system. Celiac cells are more sensitive to the gliadin peptide P31-43 and IL15 than controls. This phenotype is reproduced in control cells by inducing a delay in early vesicular trafficking. This constitutive lesion might mediate the stress/innate immune response to gliadin, which can be one of the triggers of the gliadin-specific T-cell response.


Asunto(s)
Enfermedad Celíaca/inmunología , Gliadina/inmunología , Fragmentos de Péptidos/inmunología , Adolescente , Estudios de Casos y Controles , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/patología , Niño , Preescolar , Endocitosis/inmunología , Endosomas/inmunología , Endosomas/metabolismo , Enterocitos/inmunología , Enterocitos/metabolismo , Enterocitos/patología , Receptores ErbB/metabolismo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/patología , Gliadina/metabolismo , Humanos , Inmunidad Innata , Interleucina-15/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Fragmentos de Péptidos/metabolismo , Células TH1/inmunología
11.
Sci Rep ; 8(1): 10821, 2018 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018339

RESUMEN

Celiac disease (CD) is an autoimmune disease characterized by inflammation of the intestinal mucosa due to an immune response to wheat gliadins. Some gliadin peptides are resistant to intestinal digestion (e.g., A-gliadin P31-43) and induce a stress/innate immune response, but the reason why they are dangerous in the intestines of patients with CD is unknown. In the present study, P31-43 activated IFN-α, a mediator of the innate immune response in CD, in the intestine of subjects with CD and an enterocyte cell line, CaCo-2. P31-43 cooperated with a viral ligand to activate the TLR7 pathway by interfering with endocytic trafficking. Based on these results, the vesicular pathway regulates the innate/inflammatory response to viral ligands and bioactive dietary peptides. Suggesting that together with viral infections, alimentary proteins able to mimic and potentiate the innate immune response to viruses, can trigger an autoimmune disease such as CD.


Asunto(s)
Enfermedad Celíaca/patología , Endocitosis/efectos de los fármacos , Gliadina/farmacología , Inmunidad Innata/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Adolescente , Células CACO-2 , Enfermedad Celíaca/inmunología , Niño , Preescolar , Dieta Sin Gluten , Enterocitos/citología , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Femenino , Gliadina/química , Guanosina/análogos & derivados , Guanosina/farmacología , Humanos , Interferón-alfa/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Proteínas de Resistencia a Mixovirus/metabolismo , FN-kappa B/metabolismo , Fragmentos de Péptidos/química , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
12.
Nutrients ; 9(10)2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28934109

RESUMEN

It has been hypothesized that gluten-dependent production of anti-tissue-transglutaminase 2 (anti-TG2) antibodies may occur only at an intestinal level. We have investigated intestinal production of anti-TG2 antibodies in 136 patients with normal serum levels of anti-TG2 antibodies and normal duodenal mucosa. Intestinal deposits of anti-TG2 antibodies were evaluated by immunofluorescence and anti-TG2 antibodies released in organ culture supernatants measured by ELISA. Intestinal antibody libraries were obtained from 10 subjects. Immunohistochemistry for CD25⁺, CD3⁺, and TCR-γδ⁺ was assessed in subjects with positive (n = 32) and negative (n = 31) intestinal anti-TG2 antibodies. Globally 33/136 (24%) seronegative patients produced anti-TG2 autoantibodies at an intestinal level. Antibody libraries analysis confirmed the anti-TG2 antibodies mucosal production in all (n = 8) positive subjects. Lamina propria CD25⁺ cell count was significantly (p < 0.05) higher in patients with intestinal anti-TG2. Moreover, 13/32 (41%) of them showed high TCR-γδ⁺/CD3⁺ ratios. Intestinal anti-TG2 antibody production does not show absolute specificity for CD. It is seen more often in association with inflamed mucosa. Further investigations are necessary to prove the possible role of dietary gluten.


Asunto(s)
Autoanticuerpos/análisis , Autoinmunidad , Enfermedad Celíaca/inmunología , Duodeno/inmunología , Proteínas de Unión al GTP/inmunología , Enfermedades Gastrointestinales/inmunología , Mucosa Intestinal/inmunología , Transglutaminasas/inmunología , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/enzimología , Niño , Duodeno/enzimología , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/enzimología , Humanos , Inmunidad Mucosa , Mucosa Intestinal/enzimología , Masculino , Técnicas de Cultivo de Órganos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Estudios Retrospectivos , Linfocitos T/inmunología
13.
Am J Clin Nutr ; 98(4): 1123-35, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23966426

RESUMEN

BACKGROUND: On ingestion of gliadin, the major protein component of wheat and other cereals, the celiac intestine is characterized by the proliferation of crypt enterocytes with an inversion of the differentiation/proliferation program. Gliadins and A-gliadin peptide P31-43, in particular, act as growth factors for crypt enterocytes in patients with celiac disease (CD). The effects of gliadin on crypt enterocyte proliferation and activation of innate immunity are mediated by epidermal growth factors (EGFs) and innate immunity mediators [interleukin 15 (IL15)]. OBJECTIVE: The aim of this study was to determine the molecular basis of proliferation and innate immune response to gliadin peptides in enterocytes. DESIGN: The CaCo-2 cell line was used to study EGF-, IL15-, and P31-43-induced proliferation. Silencing messenger RNAs and blocking EGF receptor and IL15 antibodies have been used to study proliferation in CaCo-2 cells and intestinal biopsy samples from patients with CD and control subjects. RESULTS: In the CaCo-2 cell model, IL15 and EGF cooperated to induce proliferation in intestinal epithelial cells at both the transcriptional and posttranscriptional levels, and the respective receptors interacted to activate each other's signaling. In addition, the effects of the P31-43 peptide on CaCo-2 cell proliferation and downstream signaling were mediated by cooperation between EGF and IL15. The increased crypt enterocyte proliferation in intestinal biopsy samples from patients with CD was reduced by EGF receptor and IL15 blocking antibodies only when used in combination. CONCLUSIONS: EGF receptor/IL15R-α cooperation regulates intestinal epithelial cell proliferation induced by EGF, IL15, and the gliadin peptide P31-43. Increased proliferation of crypt enterocytes in the intestine of CD patients is mediated by EGF/IL15 cooperation.


Asunto(s)
Enfermedad Celíaca/inmunología , Enfermedad Celíaca/patología , Enterocitos/inmunología , Enterocitos/patología , Gliadina/inmunología , Gliadina/farmacología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/farmacología , Anticuerpos/farmacología , Células CACO-2 , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Epidérmico/fisiología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Innata , Interleucina-15/genética , Interleucina-15/farmacología , Interleucina-15/fisiología , Subunidad alfa del Receptor de Interleucina-15/antagonistas & inhibidores , Subunidad alfa del Receptor de Interleucina-15/genética , Subunidad alfa del Receptor de Interleucina-15/fisiología , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transfección
14.
PLoS One ; 8(10): e76006, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204586

RESUMEN

Celiac disease (CD) occurs frequently, and is caused by ingestion of prolamins from cereals in subjects with a genetic predisposition. The small intestinal damage depends on an intestinal stress/innate immune response to certain gliadin peptides (e.g., A-gliadin P31-43) in association with an adaptive immune response to other gliadin peptides (e.g., A-gliadin P57-68). Gliadin and peptide P31-43 affect epithelial growth factor receptor (EGFR) signaling and CD enterocyte proliferation. The reason why the stress/innate immune and proliferative responses to certain gliadin peptides are present in CD and not in control intestine is so far unknown. The aim of this work is to investigate if, in CD, a constitutive alteration of enterocyte proliferation and signaling exists that may represent a predisposing condition to the damaging effects of gliadin. Immunofluorescence and immunohistochemistry were used to study signaling in CD fibroblasts and intestinal biopsies. Western blot (WB) analysis, immunoprecipitation, and quantitative PCR were also used. We found in CD enterocytes enhancement of both proliferation and Epidermal Growth Factor Receptor (EGFR)/ligand system. In CD enterocytes and fibroblasts we found increase of the phosphorylated downstream signaling molecule Extracellular Signal Regulated Kinase (ERK); block of the ERK activation normalizes enterocytes proliferation in CD mucosa. In conclusion the same pathway, which gliadin and gliadin peptide P31-43 can interfere with, is constitutively altered in CD cells. This observation potentially explains the specificity of the damaging effects of certain gliadin peptides on CD intestine.


Asunto(s)
Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/patología , Enterocitos/metabolismo , Enterocitos/patología , Transducción de Señal , Adolescente , Biopsia , Enfermedad Celíaca/genética , Proliferación Celular , Niño , Preescolar , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Lactante , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Fosforilación
15.
PLoS One ; 8(11): e79763, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278174

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca/metabolismo , Proteínas del Citoesqueleto/metabolismo , Gliadina/toxicidad , Proteínas con Dominio LIM/metabolismo , Fragmentos de Péptidos/toxicidad , Células Cultivadas , Dieta Sin Gluten/efectos adversos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Masculino
16.
PLoS One ; 6(2): e17039, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21364874

RESUMEN

BACKGROUND AND OBJECTIVES: Damage to intestinal mucosa in celiac disease (CD) is mediated both by inflammation due to adaptive and innate immune responses, with IL-15 as a major mediator of the innate immune response, and by proliferation of crypt enterocytes as an early alteration of CD mucosa causing crypts hyperplasia. We have previously shown that gliadin peptide P31-43 induces proliferation of cell lines and celiac enterocytes by delaying degradation of the active epidermal growth factor receptor (EGFR) due to delayed maturation of endocytic vesicles. IL-15 is increased in the intestine of patients affected by CD and has pleiotropic activity that ultimately results in immunoregulatory cross-talk between cells belonging to the innate and adaptive branches of the immune response. Aims of this study were to investigate the role of P31-43 in the induction of cellular proliferation and innate immune activation. METHODS/PRINCIPAL FINDINGS: Cell proliferation was evaluated by bromodeoxyuridine (BrdU) incorporation both in CaCo-2 cells and in biopsies from active CD cases and controls. We used real-time PCR to evaluate IL-15 mRNA levels and FACS as well as ELISA and Western Blot (WB) analysis to measure protein levels and distribution in CaCo-2 cells. Gliadin and P31-43 induce a proliferation of both CaCo-2 cells and CD crypt enterocytes that is dependent on both EGFR and IL-15 activity. In CaCo-2 cells, P31-43 increased IL-15 levels on the cell surface by altering intracellular trafficking. The increased IL-15 protein was bound to IL15 receptor (IL-15R) alpha, did not require new protein synthesis and functioned as a growth factor. CONCLUSION: In this study, we have shown that P31-43 induces both increase of the trans-presented IL-15/IL5R alpha complex on cell surfaces by altering the trafficking of the vesicular compartments as well as proliferation of crypt enterocytes with consequent remodelling of CD mucosa due to a cooperation of IL-15 and EGFR.


Asunto(s)
Enfermedad Celíaca/patología , Proliferación Celular/efectos de los fármacos , Gliadina/farmacología , Inmunidad Innata/efectos de los fármacos , Vesículas Transportadoras/efectos de los fármacos , Biopsia , Células CACO-2 , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/fisiopatología , Células Cultivadas , Enterocitos/inmunología , Enterocitos/metabolismo , Enterocitos/patología , Receptores ErbB/metabolismo , Receptores ErbB/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-15/genética , Interleucina-15/metabolismo , Interleucina-15/fisiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Transporte de Proteínas/efectos de los fármacos , Vesículas Transportadoras/inmunología , Vesículas Transportadoras/metabolismo
17.
PLoS One ; 5(8): e12246, 2010 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-20805894

RESUMEN

BACKGROUND: Celiac Disease (CD) is both a frequent disease (1:100) and an interesting model of a disease induced by food. It consists in an immunogenic reaction to wheat gluten and glutenins that has been found to arise in a specific genetic background; however, this reaction is still only partially understood. Activation of innate immunity by gliadin peptides is an important component of the early events of the disease. In particular the so-called "toxic" A-gliadin peptide P31-43 induces several pleiotropic effects including Epidermal Growth Factor Receptor (EGFR)-dependent actin remodelling and proliferation in cultured cell lines and in enterocytes from CD patients. These effects are mediated by delayed EGFR degradation and prolonged EGFR activation in endocytic vesicles. In the present study we investigated the effects of gliadin peptides on the trafficking and maturation of endocytic vesicles. METHODS/PRINCIPAL FINDINGS: Both P31-43 and the control P57-68 peptide labelled with fluorochromes were found to enter CaCo-2 cells and interact with the endocytic compartment in pulse and chase, time-lapse, experiments. P31-43 was localised to vesicles carrying early endocytic markers at time points when P57-68-carrying vesicles mature into late endosomes. In time-lapse experiments the trafficking of P31-43-labelled vesicles was delayed, regardless of the cargo they were carrying. Furthermore in celiac enterocytes, from cultured duodenal biopsies, P31-43 trafficking is delayed in early endocytic vesicles. A sequence similarity search revealed that P31-43 is strikingly similar to Hrs, a key molecule regulating endocytic maturation. A-gliadin peptide P31-43 interfered with Hrs correct localisation to early endosomes as revealed by western blot and immunofluorescence microscopy. CONCLUSIONS: P31-43 and P57-68 enter cells by endocytosis. Only P31-43 localises at the endocytic membranes and delays vesicle trafficking by interfering with Hrs-mediated maturation to late endosomes in cells and intestinal biopsies. Consequently, in P31-43-treated cells, Receptor Tyrosine Kinase (RTK) activation is extended. This finding may explain the role played by gliadin peptides in inducing proliferation and other effects in enterocytes from CD biopsies.


Asunto(s)
Gliadina/metabolismo , Fragmentos de Péptidos/metabolismo , Vesículas Transportadoras/metabolismo , Secuencia de Aminoácidos , Animales , Biopsia , Células CACO-2 , Ciclo Celular , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/metabolismo , Enterocitos/metabolismo , Receptores ErbB/metabolismo , Gliadina/química , Humanos , Intestino Delgado/patología , Ratones , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Transporte de Proteínas , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA