Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 10798, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612145

ABSTRACT

Celiac disease (CD) is an enteropathy triggered by the ingestion of gluten proteins in genetically predisposed individuals and characterized by excessive activation of effector immune cells and enhanced production of inflammatory cytokines. However, factors/mechanisms that amplify the ongoing mucosal inflammation in CD are not fully understood. In this study, we assessed whether mammalian target of Rapamycin (mTOR), a pathway that combines intra- and extra-cellular signals and acts as a central regulator for the metabolism, growth, and function of immune and non-immune cells, sustains CD-associated immune response. Our findings indicate that expression of phosphorylated (p)/active form of mTOR is increased in protein lysates of duodenal biopsy samples taken from patients with active CD (ACD) as compared to normal controls. In ACD, activation of mTOR occurs mainly in the epithelial compartment and associates with enhanced expression of p-4EBP, a downstream target of mTOR complex (mTORC)1, while expression of p-Rictor, a component of mTORC2, is not increased. Stimulation of mucosal explants of inactive CD patients with pepsin-trypsin-digested (PT)-gliadin or IFN-γ/IL-21, two cytokines produced in CD by gluten-specific T cells, increases p-4EBP expression. Consistently, blockade of such cytokines in cultures of ACD mucosal explants reduces p-4EBP. Finally, we show that inhibition of mTORC1 with rapamycin in ACD mucosal explants reduces p-4EBP and production of IL-15, a master cytokine produced by epithelial cells in this disorder. Our data suggest that ACD inflammation is marked by activation of mTORC1 in the epithelial compartment.


Subject(s)
Celiac Disease/immunology , Duodenum/immunology , Intestinal Mucosa/immunology , TOR Serine-Threonine Kinases/immunology , Biopsy , Celiac Disease/pathology , Duodenum/pathology , Female , Gliadin/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/immunology , Interleukins/immunology , Intestinal Mucosa/pathology , Male , Mechanistic Target of Rapamycin Complex 1/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Phosphorylation/immunology , T-Lymphocytes/immunology
2.
Aliment Pharmacol Ther ; 40(8): 974-81, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25112824

ABSTRACT

BACKGROUND: The clinical presentation of organic and functional intestinal disorders can overlap and clinicians often rely on invasive and time-consuming procedures to make a final diagnosis. Regenerating islet-derived 3-alpha (Reg3α) is detectable in the circulation of patients with intestinal graft-versus host disease and patients with inflammatory bowel disease (IBD). AIM: To determine whether serum Reg3α testing is useful for discriminating mucosal enteropathies from functional intestinal disorders. METHODS: We prospectively included 47 patients with active coeliac disease (ACD), 13 patients with refractory coeliac disease (RCD), seven patients with common variable immunodeficiency (CVID), 72 patients with active Crohn's disease, 22 patients with active ulcerative colitis (UC) and 28 patients with irritable bowel syndrome (IBS)-related diarrhoea. Sera were also taken from 10 CD patients before and after 6-12 months of a gluten-free diet (GFD) and from 14 patients with IBD before and after induction therapy with Infliximab (IFX). Sera of 119 healthy volunteers were used to determine the cut-off value. Reg3α levels were measured by a commercial ELISA kit. RESULTS: Levels of Reg3α exceeded the cut-off value of the assay in 43/47(91%) ACD patients, 13/13(100%) RCD patients, 7/7(100%) CVID patients, 65/72(90%) Crohn's disease patients, 17/22(77%) UC patients and one patient with IBS(4%). Reg3α levels distinguished mucosal enteropathies from IBS with a sensitivity of 90% and a specificity of 96%. Reg3α levels significantly decreased in CD patients following a GFD and in IBD patients after treatment with IFX. CONCLUSION: Reg3α is a serum biomarker of intestinal damage that, combined with clinical data, identifies patients who should undergo invasive tests for diagnosing enteropathies.


Subject(s)
Antigens, Neoplasm/blood , Biomarkers, Tumor/blood , Celiac Disease/blood , Colitis, Ulcerative/blood , Common Variable Immunodeficiency/blood , Crohn Disease/blood , Irritable Bowel Syndrome/blood , Lectins, C-Type/blood , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Celiac Disease/diagnosis , Colitis, Ulcerative/diagnosis , Common Variable Immunodeficiency/diagnosis , Crohn Disease/diagnosis , Female , Humans , Irritable Bowel Syndrome/diagnosis , Male , Middle Aged , Pancreatitis-Associated Proteins , Young Adult
3.
Mucosal Immunol ; 7(6): 1467-79, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24850427

ABSTRACT

In inflammatory bowel disease (IBD), tissue damage is driven by an excessive immune response, poorly controlled by counter-regulatory mechanisms. SIRT1, a class III NAD+-dependent deacetylase, regulates negatively the expression of various proteins involved in the control of immune-inflammatory pathways, such as Stat3, Smad7, and NF-κB. Here we examined the expression, regulation, and function of SIRT1 in IBD. SIRT1 RNA and protein expression was less pronounced in whole biopsies and lamina propria mononuclear cells (LPMCs) of IBD patients in comparison with normal controls. SIRT1 expression was downregulated in control LPMC by tumor necrosis factor (TNF)-α and interleukin (IL)-21, and upregulated in IBD LPMC by neutralizing TNF-α and IL-21antibodies. Consistently, SIRT1 expression was increased in mucosal samples taken from IBD patients successfully treated with Infliximab. Treatment of IBD LPMC with Cay10591, a specific SIRT1 activator, reduced NF-κB activation and inhibited inflammatory cytokine synthesis, whereas Ex527, an inhibitor of SIRT1, increased interferon (IFN)-γ in control LPMC. SIRT1 was also reduced in mice with colitis induced by 2,4,6-trinitrobenzenesulphonic acid or oxazolone. Cay10591 prevented and cured experimental colitis whereas Ex527 exacerbated disease by modulating T cell-derived cytokine response. Data indicate that SIRT1 is downregulated in IBD patients and colitic mice and suggest that SIRT1 activation can help attenuate inflammatory signals in the gut.


Subject(s)
Gene Expression Regulation, Enzymologic/immunology , Inflammatory Bowel Diseases/immunology , Intestines/immunology , Sirtuin 1/immunology , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/pharmacology , Adult , Aged , Aged, 80 and over , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Antibodies, Monoclonal/administration & dosage , Female , Gene Expression Regulation, Enzymologic/genetics , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Infliximab , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukins/genetics , Interleukins/immunology , Intestines/pathology , Male , Mice , Middle Aged , Oxazolone/adverse effects , Oxazolone/pharmacology , Sirtuin 1/genetics , Trinitrobenzenesulfonic Acid/toxicity , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...