Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Clin Dev Immunol ; 2012: 329150, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22899947

RESUMEN

A lifelong gluten-free diet (GFD) is mandatory for celiac disease (CD) but has poor compliance, justifying novel strategies. We found that wheat flour transamidation inhibited IFN-γ secretion by intestinal T cells from CD patients. Herein, the primary endpoint was to evaluate the ability of transamidated gluten to maintain GFD CD patients in clinical remission. Secondary endpoints were efficacy in prevention of the inflammatory response and safety at the kidney level, where reaction products are metabolized. In a randomized single blinded, controlled 90-day trial, 47 GFD CD patients received 3.7 g/day of gluten from nontransamidated (12) or transamidated (35) flour. On day 15, 75% and 37% of patients in the control and experimental groups, respectively, showed clinical relapse (P = 0.04) whereas intestinal permeability was mainly altered in the control group (50% versus 20%, P = 0.06). On day 90, 0 controls and 14 patients in the experimental group completed the challenge with no variation of antitransglutaminase IgA (P = 0.63), Marsh-Oberhuber grading (P = 0.08), or intestinal IFN-γ mRNA (P > 0.05). Creatinine clearance did not vary after 90 days of treatment (P = 0.46). In conclusion, transamidated gluten reduced the number of clinical relapses in challenged patients with no changes of baseline values for serological/mucosal CD markers and an unaltered kidney function.


Asunto(s)
Enfermedad Celíaca/dietoterapia , Dieta Sin Gluten , Proteínas en la Dieta/administración & dosificación , Harina , Glútenes/administración & dosificación , Triticum/metabolismo , Adolescente , Adulto , Amidinotransferasas/inmunología , Autoanticuerpos/sangre , Enfermedad Celíaca/inmunología , Ingestión de Alimentos , Femenino , Humanos , Interferón gamma/metabolismo , Mucosa Intestinal/inmunología , Masculino , Persona de Mediana Edad , Cooperación del Paciente , Método Simple Ciego , Transglutaminasas/metabolismo , Triticum/química , Adulto Joven
2.
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
3.
BMC Med ; 9: 23, 2011 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-21392369

RESUMEN

BACKGROUND: Celiac disease (CD) is an autoimmune enteropathy triggered by the ingestion of gluten. Gluten-sensitive individuals (GS) cannot tolerate gluten and may develop gastrointestinal symptoms similar to those in CD, but the overall clinical picture is generally less severe and is not accompanied by the concurrence of tissue transglutaminase autoantibodies or autoimmune comorbidities. By studying and comparing mucosal expression of genes associated with intestinal barrier function, as well as innate and adaptive immunity in CD compared with GS, we sought to better understand the similarities and differences between these two gluten-associated disorders. METHODS: CD, GS and healthy, gluten-tolerant individuals were enrolled in this study. Intestinal permeability was evaluated using a lactulose and mannitol probe, and mucosal biopsy specimens were collected to study the expression of genes involved in barrier function and immunity. RESULTS: Unlike CD, GS is not associated with increased intestinal permeability. In fact, this was significantly reduced in GS compared with controls (P = 0.0308), paralleled by significantly increased expression of claudin (CLDN) 4 (P = 0.0286). Relative to controls, adaptive immunity markers interleukin (IL)-6 (P = 0.0124) and IL-21 (P = 0.0572) were expressed at higher levels in CD but not in GS, while expression of the innate immunity marker Toll-like receptor (TLR) 2 was increased in GS but not in CD (P = 0.0295). Finally, expression of the T-regulatory cell marker FOXP3 was significantly reduced in GS relative to controls (P = 0.0325) and CD patients (P = 0.0293). CONCLUSIONS: This study shows that the two gluten-associated disorders, CD and GS, are different clinical entities, and it contributes to the characterization of GS as a condition associated with prevalent gluten-induced activation of innate, rather than adaptive, immune responses in the absence of detectable changes in mucosal barrier function.


Asunto(s)
Enfermedad Celíaca/inmunología , Enfermedad Celíaca/patología , Hipersensibilidad/inmunología , Hipersensibilidad/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Permeabilidad , Adulto , Alérgenos/inmunología , Femenino , Perfilación de la Expresión Génica , Glútenes/inmunología , Humanos , Masculino
4.
Scand J Gastroenterol ; 46(10): 1194-205, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21843037

RESUMEN

OBJECTIVE: Most of the recent studies suggest that oats are well tolerated by celiac disease (CD) patients. However, it is still possible that different oat cultivars may display different biological properties relevant for CD pathogenesis. We aimed to investigate biological and immunological properties of two oat varieties, Avena genziana and Avena potenza, in relation to their safety for CD patients. MATERIAL AND METHODS: Phosphorylation of extracellular signal-regulated kinase (ERK) and trans-epithelial electrical resistance (TEER) were evaluated in CaCo-2 cells treated with peptic-tryptic (PT) digests from the two oats and from gliadin (PTG). With the same PT-digests, duodenal biopsies from 22 CD patients were treated in vitro for 24 h and density of CD25+ cells in lamina propria and of intraepithelial CD3+ T cells was measured, as well as crypt cell proliferation and epithelial expression of interleukin 15. Finally, interferon γ (IFN-γ) production was measured as evidence of gliadin-specific T-cell activation by PT-digests. RESULTS: In contrast to PTG, oats PT-digests were not able to induce significant increase in ERK phosphorylation and decrease in TEER in CaCo-2 cells. In the organ culture system, oats PT-digests, unlike PTG, did not induce significant increase in crypt enterocyte proliferation, increase in interleukin 15 expression or in lamina propria CD25+ cells. Nevertheless Avena potenza increased intraepithelial T-cell density, while Avena genziana-induced IFN-γ production in 3/8 CD intestinal T cell lines. CONCLUSIONS: Our data show that Avena genziana and Avena potenza do not display in vitro activities related to CD pathogenesis. Some T-cell reactivity could be below the threshold for clinical relevance.


Asunto(s)
Avena/efectos adversos , Avena/inmunología , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/patología , Adolescente , Adulto , Biopsia , Complejo CD3/metabolismo , Células CACO-2 , Proliferación Celular , Niño , Preescolar , Impedancia Eléctrica , Enterocitos/citología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Gliadina/inmunología , Humanos , Interferón gamma/metabolismo , Interleucina-15/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Activación de Linfocitos/inmunología , Persona de Mediana Edad , Fosforilación , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto Joven
5.
Eur J Immunol ; 39(12): 3552-61, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19795413

RESUMEN

Celiac disease (CD) is an enteropathy triggered by gluten and mediated by CD4+ T cells. A complete understanding of CD immunopathogenesis has been hindered due to the lack of adequate in vivo models. Here, we explored the effect of the inhibition of COX by indomethacin in wheat gliadin-sensitized transgenic mice expressing the HLA-DQ8 heterodimer, a molecule associated with CD. Treated mice showed a gliadin-specific immune response with a significant reduction of villus height, not linked to crypt hyperplasia and to expansion of intraepithelial T cells. Notably, treated mice showed increased numbers of CD25+ and apoptotic cells in the lamina propria, whereas high basal levels of IFN-gamma secretion, along with a reduced gliadin-specific IL-2 expression were detected in MLN. Biochemical assessment of the lesion revealed increased mRNA of Lamb3 and Adamts2, encoding for ECM proteins, and enhanced activities of metalloproteinases MMP1, 2 and 7. We conclude that an intestinal sensitivity to gliadin, in connection with COX inhibition, caused a decreased villus height in DQ8 tg mice. The lesion was induced by a deregulated mucosal cell immunity to gliadin, thus triggering activation of a specific ECM protein pathway responsible for lamina propria remodeling.


Asunto(s)
Enfermedad Celíaca/inmunología , Gliadina/inmunología , Antígenos HLA-DQ/inmunología , Mucosa Intestinal/inmunología , Proteínas ADAM/genética , Proteínas ADAMTS , Proteína ADAMTS4 , Animales , Apoptosis , Complejo CD3/metabolismo , Enfermedad Celíaca/genética , Enfermedad Celíaca/metabolismo , Moléculas de Adhesión Celular/genética , Inhibidores de la Ciclooxigenasa/farmacología , Factores de Transcripción Forkhead/metabolismo , Antígenos HLA-DQ/genética , Humanos , Inmunohistoquímica , Indometacina/farmacología , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Procolágeno N-Endopeptidasa/genética , Prostaglandina-Endoperóxido Sintasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Kalinina
6.
Gastroenterology ; 134(4): 1017-27, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18395083

RESUMEN

BACKGROUND & AIMS: The extensive infiltration of CD8(+) T cells in the intestinal mucosa of celiac disease (CD) patients is a hallmark of the disease. We identified a gliadin peptide (pA2) that is selectively recognized by CD8(+) T cells infiltrating intestinal mucosa of HLA-A2(+) CD patients. Herein, we investigated the phenotype, the tissue localization, and the effector mechanism of cells responsive to pA2 by using the organ culture of CD intestinal mucosa. The target of pA2-mediated cytotoxicity was also investigated by using the intestinal epithelial cell lines Caco2 and HT29, A2(+) and A2(-), respectively, as target cells. METHODS: Jejunal biopsy specimens from CD patients were cultured in vitro with pA2, and cellular activation was evaluated by immunohistochemistry and cytofluorimetric analysis. Cytotoxicity of pA2-specific, intestinal CD8(+) T cells was assayed by granzyme-B and interferon-gamma release and by apoptosis of target cells. RESULTS: pA2 challenge of A2(+) CD mucosa increased the percentage of CD8(+)CD25(+) and of CD80(+) cells in the lamina propria, the former mainly localized beneath the epithelium, as well as the number of terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling-positive cells (TUNEL(+)) in the epithelium. Intraepithelial CD3(+) cells and enterocyte expression of Fas were also increased. CD8(+)CD25(+) and CD8(+)FASL(+) T cells were significantly increased in cell preparations from biopsy specimens cultured with pA2. CD8(+) T-cell lines released both granzyme-B and interferon-gamma following recognition of pA2 when presented by Caco2 and not by HT29. CONCLUSIONS: These data indicate that gliadins contain peptides able to activate, through a TCR/HLA class I interaction, CD8-mediated response in intestinal CD mucosa and to induce the enterocyte apoptosis.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedad Celíaca/inmunología , Enterocitos/patología , Gliadina/metabolismo , Antígeno HLA-A2/inmunología , Yeyuno/patología , Activación de Linfocitos/inmunología , Adulto , Antígeno B7-1/inmunología , Biopsia , Linfocitos T CD8-positivos/metabolismo , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/patología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Gliadina/efectos adversos , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Masculino , Persona de Mediana Edad , Técnicas de Cultivo de Órganos , Receptor fas/inmunología
7.
Immunol Lett ; 119(1-2): 78-83, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18547649

RESUMEN

Probiotic strains have been reported to exert immunomodulatory activities in the gut-associated lymphoid tissue. In this study we explored the effect of Lactobacillus casei in transgenic mice expressing the human DQ8 heterodimer, a HLA molecule linked to Celiac Disease (CD). DQ8 mice, mucosally immunized with the gluten component gliadin, mounted an intestinal Th1-like response as observed in CD, without developing enteropathy. Co-administration of L. casei in sensitized mice specifically enhanced the gliadin-specific response mediated by CD4(+) T cells. Notably, both a strong increase of the gliadin-specific IFNgamma expression and a pro-inflammatory polarization of the cytokine milieu in the small intestinal mucosa were associated to the presence of the probiotic strain. However, this condition did not bring on any mucosal alteration. These findings suggest that the gliadin-specific enteropathy is not merely related to the HLA DQ8-restricted massive production of IFNgamma, but additional parameters are involved. Moreover, our data imply that the intrinsic adjuvanticity of L. casei can be exploited to further enhance both mucosal and systemic T cell-mediated responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos Bacterianos/inmunología , Enfermedad Celíaca/microbiología , Inmunidad Mucosa/inmunología , Lacticaseibacillus casei/inmunología , Células TH1/metabolismo , Animales , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/terapia , Proliferación Celular/efectos de los fármacos , Gliadina/administración & dosificación , Glútenes/inmunología , Antígenos HLA-DQ/inmunología , Antígenos HLA-DQ/metabolismo , Humanos , Tolerancia Inmunológica/inmunología , Inmunidad Mucosa/efectos de los fármacos , Inmunización , Interferón gamma/agonistas , Interferón gamma/metabolismo , Interleucina-10/antagonistas & inhibidores , Interleucina-10/metabolismo , Intestino Delgado/inmunología , Lacticaseibacillus casei/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Células TH1/citología , Células TH1/efectos de los fármacos , Hipersensibilidad al Trigo/inmunología
8.
Res Microbiol ; 157(9): 891-7, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17005378

RESUMEN

The present work was aimed at investigating whether Bacillus subtilis spores, widely used in probiotic as well as pharmaceutical preparations for mild gastrointestinal disorders, can suppress enteric infections. To address this issue, we developed a mouse model of infection using the mouse enteropathogen Citrobacter rodentium, a member of a family of human and animal pathogens which includes the clinically significant enteropathogenic (EPEC) and enterohemorrhagic (EHEC) Escherichia coli strains. This group of pathogens causes transmissible colonic hyperplasia by using attaching and effacing (A/E) lesions to colonize the host colon. Because of its similarities to human enteropathogens, C. rodentium is now widely used as an in vivo model for gastrointestinal infections. Swiss NIH mice were orally administered B. subtilis spores one day before infection with C. rodentium. Mice were sacrificed on day 15 after infection, and distal colon, liver and mesenteric lymph nodes were removed for bacteria counts, morphology, immunohistology and IFNgamma mRNA analysis. We observed that spore predosing was effective in significantly decreasing infection and enteropathy in suckling mice infected with a dose of C. rodentium sufficient to cause colon colonization, crypt hyperplasia and high mortality rates. Moreover, in mice predosed with spores, the number of CD4(+) cells and IFNgamma transcript levels remained high. These results thus indicate that our newly established model of C. rodentium infection is a suitable system for analyzing the effects of probiotic bacteria on enteroinfections and that B. subtilis spores are efficient at reducing C. rodentium infection in mice, leaving unaltered the immune response against the pathogen.


Asunto(s)
Bacillus subtilis/fisiología , Citrobacter rodentium/crecimiento & desarrollo , Susceptibilidad a Enfermedades/microbiología , Enfermedades Intestinales/microbiología , Animales , Bacillus subtilis/crecimiento & desarrollo , Antígenos CD4/análisis , Modelos Animales de Enfermedad , Expresión Génica/genética , Inmunohistoquímica , Interferón gamma/genética , Enfermedades Intestinales/genética , Enfermedades Intestinales/metabolismo , Ratones , Esporas Bacterianas/crecimiento & desarrollo , Esporas Bacterianas/fisiología
9.
Mol Nutr Food Res ; 55 Suppl 2: S248-56, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21954188

RESUMEN

SCOPE: The involvement of oxidative stress in gluten-induced toxicity has been evidenced in vitro and in clinical studies but has never been examined in vivo. We recently demonstrated the protective activity of conjugated linoleic acid (CLA), which functions by the activation of nuclear factor erythroid 2-related factor2 (Nrf2), a key transcription factor for the synthesis of antioxidant and detoxifying enzymes (phase 2). Here, we evaluate the involvement of nuclear factor erythroid 2-related factor2 in gliadin-mediated toxicity in human Caco-2 intestinal cells and in gliadin-sensitive human leukocyte antigen-DQ8 transgenic mice (DQ8) and the protective activity of CLA. METHODS AND RESULTS: Gliadin effects in differentiated Caco-2 cells and in DQ8 mice, fed with a gliadin-containing diet with or without CLA supplementation, were evaluated by combining enzymatic, immunochemical, immunohistochemical, and quantitative real-time PCR (qRT-PCR) assays. Gliadin toxicity was accompanied by downregulation of phase 2 and elevates proteasome-acylpeptide hydrolase activities in vitro and in vivo. Notably, gliadin was unable to generate severe oxidative stress extent or pathological consequences in DQ8 mice intestine comparable to those found in celiac patients and the alterations produced were hampered by CLA. CONCLUSION: The beneficial effects of CLA against the depletion of crucial intestinal cytoprotective defenses indicates a novel nutritional approach for the treatment of intestinal disease associated with altered redox homeostasis.


Asunto(s)
Gliadina/toxicidad , Intestinos/efectos de los fármacos , Intestinos/fisiología , Ácidos Linoleicos Conjugados/farmacología , Animales , Antioxidantes/metabolismo , Células CACO-2 , Enfermedad Celíaca/metabolismo , Diferenciación Celular/efectos de los fármacos , Enzimas/genética , Enzimas/metabolismo , Gliadina/efectos adversos , Antígenos HLA-DQ/genética , Humanos , Inactivación Metabólica , Ratones , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Transglutaminasas/metabolismo
10.
J Lipid Res ; 47(11): 2382-91, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16877747

RESUMEN

Conjugated linoleic acid (CLA), a naturally occurring peroxisome proliferator-activated receptor gamma (PPAR gamma) ligand, exhibits proapoptotic, immunomodulatory, and anticancer properties. In this study, we examined the biological effects of CLA administration in the MRL/MpJ-Fas(lpr) mouse, an animal model of systemic lupus erythematosus (SLE). We found that CLA exerted apparently opposed activities in in vitro experiments, depending on its concentration: 100 microM CLA downregulated IFN gamma synthesis and cell proliferation of splenocytes, in association with apoptosis induction and a decrease of intracellular thiols (GSH + GSSG), whereas 25 microM CLA did not significantly influence cell proliferation but enhanced the expression of gamma-glutamylcysteine ligase catalytic subunit (GCLC) and intracellular GSH concentration. Interestingly, the antiproliferative effect at 100 microM was not inhibited by the PPAR gamma antagonist GW9662. In vivo, CLA administration drastically reduced SLE signs (splenomegaly, autoantibodies, and cytokine synthesis), a condition paralleled by the enhancement of GCLC expression and intracellular GSH content. Moreover, CLA administration significantly downregulated nuclear factor kappaB activity independent of PPAR gamma activation and apoptosis induction. In conclusion, enhanced GSH content and GCLC expression in CLA-treated mice suggest a novel biochemical mechanism underlying its immunomodulatory activity and the beneficial effects on murine SLE signs.


Asunto(s)
Glutatión/metabolismo , Ácidos Linoleicos Conjugados/farmacología , Anilidas/farmacología , Animales , Apoptosis , Proliferación Celular , Citocinas/metabolismo , Regulación hacia Abajo , Femenino , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos MRL lpr , FN-kappa B/metabolismo , Tamaño de los Órganos , PPAR gamma/metabolismo , Especies Reactivas de Oxígeno , Bazo/citología , Bazo/embriología , Bazo/metabolismo
11.
Am J Pathol ; 162(6): 1845-55, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12759242

RESUMEN

The biological effects of interferon (IFN)-gamma rely mainly on the activity of the transcription factor signal transducer and activator of transcription (STAT) 1 and the intracellular levels of suppressor of cytokine signaling (SOCS)-1, a negative regulator that controls the amplitude and duration of STAT-1 activation. IFN-gamma is a key mediator of the immunopathology in celiac disease (CD, gluten-sensitive enteropathy). Thus we have investigated STAT-1 signaling and SOCS-1 expression in this condition. As expected, high local concentrations of IFN-gamma were invariably seen in duodenal biopsies from CD patients in comparison to controls. On the basis of immunohistochemistry, STAT-1 phosphorylation, nuclear localization, and DNA-binding activity, STAT-1 activation was consistently more pronounced in CD compared with controls. Despite samples from CD patients containing abundant SOCS-1 mRNA, SOCS-1 protein was expressed at the same level in CD patients and controls. In explant cultures of CD biopsies, gliadin induced the activation of STAT-1 but not SOCS-1. Furthermore, inhibition of STAT-1 prevented the gliadin-mediated induction of ICAM-1 and B7-2. These data suggest that persistent STAT-1 activation can contribute to maintaining and expanding the local inflammatory response in CD.


Asunto(s)
Enfermedad Celíaca/patología , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas Represoras , Transactivadores/metabolismo , Factores de Transcripción , Adolescente , Adulto , Antígenos CD/metabolismo , Antígeno B7-2 , Western Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Enfermedad Celíaca/genética , Enfermedad Celíaca/metabolismo , Células Cultivadas , Niño , Preescolar , Expresión Génica , Gliadina/farmacología , Humanos , Inmunohistoquímica , Molécula 1 de Adhesión Intercelular/metabolismo , Interferón gamma/metabolismo , Interferón gamma/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Leucocitos Mononucleares/metabolismo , Glicoproteínas de Membrana/metabolismo , Fosforilación , Proteínas/genética , Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA