Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mariantonia Maglio is active.

Publication


Featured researches published by Mariantonia Maglio.


Clinical Gastroenterology and Hepatology | 2011

Natural History of Potential Celiac Disease in Children

A. Tosco; V.M. Salvati; Renata Auricchio; Mariantonia Maglio; Melissa Borrelli; A. Coruzzo; Francesco Paparo; Massimo Boffardi; Antonella Esposito; Grazia D'Adamo; B. Malamisura; Luigi Greco; Riccardo Troncone

BACKGROUND & AIMS The presence of celiac disease-associated autoantibodies (antiendomysium and antitissue transglutaminase [anti-TG2]) with normal jejunal mucosa indicate potential celiac disease. We performed a prospective, 3-year cohort study to determine the natural history of potential celiac disease in children. METHODS The study included 106 children with potential celiac disease, based on serology analysis and normal duodenal architecture. All but 2 carried the HLA-DQ2 and/or DQ8 haplotype. In all children, every 6 months, growth, nutritional parameters, celiac disease serology, and autoimmunity were investigated. In biopsies, γδ intraepithelial-, CD3-, and lamina propria CD25-positive cells were counted; duodenal deposits of anti-TG2 immunoglobulin A were detected. Biopsy analysis was repeated after 2 years on patients with persistent positive serology and/or symptoms. RESULTS Celiac disease was detected primarily in first-degree relatives and patients with autoimmune disorders (40.6%). A gluten-free diet was prescribed to 20/106 patients because of symptoms, which were relieved in only 11. Eighty-nine of the 106 patients entered the follow-up study, with normal daily consumption of gluten. During the follow-up antibodies disappeared in 14.6% and fluctuated in 32.6%. Villous atrophy was observed in 12/39 patients (30.8%) who underwent a repeat biopsy. CONCLUSIONS Most children with potential celiac disease remain healthy. After 3 years, approximately 33% of patients develop villous atrophy. Intestinal deposits of anti-TG2 IgA identify children at risk for villous atrophy.


PLOS ONE | 2011

Gliadin-Mediated Proliferation and Innate Immune Activation in Celiac Disease Are Due to Alterations in Vesicular Trafficking

M. Vittoria Barone; Delia Zanzi; Mariantonia Maglio; Merlin Nanayakkara; S. Santagata; Giuliana Lania; Erasmo Miele; Maria T. Ribecco; Francesco Maurano; Renata Auricchio; Carmen Gianfrani; Silvano Ferrini; Riccardo Troncone; Salvatore Auricchio

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.


The American Journal of Gastroenterology | 2014

Potential Celiac Children: 9-Year Follow-Up on a Gluten-Containing Diet

Renata Auricchio; A. Tosco; Emanuela Piccolo; Martina Galatola; Valentina Izzo; Mariantonia Maglio; Francesco Paparo; Riccardo Troncone; Luigi Greco

OBJECTIVES:Potential celiac disease (CD) is defined by the presence of serum anti-tissue-transglutaminase (anti-TG2) antibodies and normal duodenal mucosa. The major clinical problem is the management of asymptomatic patients and how to predict the development of villous atrophy. This prospective longitudinal cohort study describes the natural history of potential CD up to 9 years and explores risk factors associated with the development of mucosal damage.METHODS:Two hundred and ten potential CD children were eligible for the study; 175/210 asymptomatic children were left on a gluten-containing diet. Antibodies and clinical symptoms were checked every 6 months, and a small bowel biopsy was taken every 2 years to evaluate histological, immunohistochemical, and anti-TG2 deposits. Patients were genotyped for HLA and a set of non-HLA CD-associated genes.RESULTS:Forty-three percent of patients showed persistently elevated anti-TG2 level, 20% became negative during follow-up, and 37% showed a fluctuant anti-TG2 course with transiently negative values. At 3 years of follow-up, 86% of cases remained potential; 73 and 67% still had normal duodenal architecture at 6 and 9 years, respectively. Male sex, slight mucosal inflammation at time 0, and a peculiar genetic profile delineate a cohort of individuals who were prone to develop mucosal damage during time.CONCLUSIONS:A sizeable proportion of asymptomatic potential celiac patients showed fluctuation or negativization of antibody production, and many of these, with persistently positive anti-TG2, did not develop mucosal damage after 9 years of follow-up. Celiac population is a multivariate aggregate of individuals with different genetic and phenotypic profiles. Caution is required before prescribing a gluten-free diet for life to asymptomatic individuals with potential CD.


PLOS ONE | 2010

Gliadin Peptide P31-43 Localises to Endocytic Vesicles and Interferes with Their Maturation

Maria Vittoria Barone; Merlin Nanayakkara; Giovanni Paolella; Mariantonia Maglio; Virginia Vitale; Raffaele Troiano; Maria T. Ribecco; Giuliana Lania; Delia Zanzi; S. Santagata; Renata Auricchio; Riccardo Troncone; Salvatore Auricchio

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 Tyrosin 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.


Diabetes | 2013

Promotion of Autoimmune Diabetes by Cereal Diet in the Presence or Absence of Microbes Associated With Gut Immune Activation, Regulatory Imbalance, and Altered Cathelicidin Antimicrobial Peptide

Christopher Patrick; Gen-Sheng Wang; David E. Lefebvre; Jennifer A. Crookshank; Brigitte Sonier; Chandra Eberhard; Majid Mojibian; Chris R. J. Kennedy; Stephen P. J. Brooks; Martin Kalmokoff; Mariantonia Maglio; Riccardo Troncone; Philippe Poussier; Fraser W. Scott

We are exposed to millions of microbial and dietary antigens via the gastrointestinal tract, which likely play a key role in type 1 diabetes (T1D). We differentiated the effects of these two major environmental factors on gut immunity and T1D. Diabetes-prone BioBreeding (BBdp) rats were housed in specific pathogen-free (SPF) or germ-free (GF) conditions and weaned onto diabetes-promoting cereal diets or a protective low-antigen hydrolyzed casein (HC) diet, and T1D incidence was monitored. Fecal microbiota 16S rRNA genes, immune cell distribution, and gene expression in the jejunum were analyzed. T1D was highest in cereal-SPF (65%) and cereal-GF rats (53%) but inhibited and delayed in HC-fed counterparts. Nearly all HC-GF rats remained diabetes-free, whereas HC-fed SPF rats were less protected (7 vs. 29%). Bacterial communities differed in SPF rats fed cereal compared with HC. Cereal-SPF rats displayed increased gut CD3+ and CD8α+ lymphocytes, ratio of Ifng to Il4 mRNA, and Lck expression, indicating T-cell activation. The ratio of CD3+ T cells expressing the Treg marker Foxp3+ was highest in HC-GF and lowest in cereal-SPF rats. Resident CD163+ M2 macrophages were increased in HC-protected rats. The cathelicidin antimicrobial peptide (Camp) gene was upregulated in the jejunum of HC diet–protected rats, and CAMP+ cells colocalized with CD163. A cereal diet was a stronger promoter of T1D than gut microbes in association with impaired gut immune homeostasis.


Journal of Pediatric Gastroenterology and Nutrition | 2010

Serum and intestinal celiac disease-associated antibodies in children with celiac disease younger than 2 years of age.

Mariantonia Maglio; A. Tosco; Francesco Paparo; Renata Auricchio; Viviana Granata; B. Colicchio; Viviana Indolfi; Erasmo Miele; Riccardo Troncone

Objectives: In children younger than 2 years of age, a diagnosis of celiac disease (CD) is difficult to make because anti-endomysium (anti-EMA)/anti-tissue transglutaminase 2 (anti-TG2) antibodies are less sensitive than in older children. The aim of our study was to evaluate how many children younger than 2 years of age and diagnosed with CD, were negative for serum anti-TG2 antibodies and to test the hypothesis that in these patients, TG2-specific IgA deposits could instead be present at mucosal level. Patients and Methods: A total of 104 children younger than 2 years of age and 179 children older than 2 years, all of whom had been diagnosed with CD, were investigated for serum CD-associated antibodies (anti-gliadin [AGA] IgA and IgG, EMA-IgA, anti-TG2–IgA). The presence of intestinal anti-TG2 extracellular IgA deposits was searched by using double immunofluorescence in 56 of the patients younger than 2 years of age and in 40 of those who were older than 2 years. Results: In children with CD who were younger than 2 years of age, high levels of AGA-IgA were found in 93/104 (89%) and 98/104 (94%) were found of have high levels of AGA-IgG. In children older than the age of 2 years with CD, 120/179 (67%) had high levels of AGA-IgA and 151/179 (84%) had high levels of AGA-IgG. Serum EMA were present in 92/104 (88%) in the younger group and in 176/179 (98%) of the older group. Ninety-one of 104 children (87%) younger and 172/179 (96%) older than 2 years showed high serum levels of anti-TG2. Finally, 41/56 (73%) children younger than 2 years and all of the 40 children (100%) older than 2 years of age showed mucosal anti-TG2–IgA deposits. Conclusions: EMA and anti-TG2–antibody measurements show higher sensitivity for the diagnosis of CD in children older than 2 years compared with younger children. The search for mucosal deposits of anti-TG2–IgA does not improve the diagnostic performance.


Digestive and Liver Disease | 2011

Intestinal deposits of anti-tissue transglutaminase IgA in childhood celiac disease

Mariantonia Maglio; A. Tosco; Renata Auricchio; Francesco Paparo; B. Colicchio; Erasmo Miele; L. Rapacciuolo; Riccardo Troncone

BACKGROUND High serum levels of anti-tissue-transglutaminase-2 IgA antibodies (anti-TG2), which are produced and deposited in the intestine, characterize celiac disease. AIM To assess the diagnostic value of intestinal deposits of anti-TG2 IgA for celiac disease in a paediatric population. METHODS 344 children underwent duodenal biopsy for the suspicion of CD, and were divided into 3 groups: group A, 144 celiac subjects with villous atrophy (Marsh 3b-c); group B, 109 subjects with high serum levels of anti-TG2 but normal intestinal mucosa (Marsh 0-1) (potential celiac disease patients); group C, 91 subjects with normal levels of serum anti-TG2: 70 with Marsh 0-1 and 21 with Marsh 3a mucosa. All duodenal sections were evaluated for the presence of intestinal deposits of anti-TG2 IgA by double immunofluorescence. RESULTS Deposits of anti-TG2 IgA were present in 96%, 68%, 12% of patients from groups A, B, C, respectively. Diagnostic sensitivity and specificity for celiac disease were 96% and 88% vs. 97% and 100% for serum anti-TG2, respectively. The degree of concordance with serum anti-TG2 was 85%. CONCLUSION Detection of intestinal deposits of anti-TG2 IgA is a useful diagnostic tool. Further research is needed regarding their ability to predict evolution to villous atrophy in potential celiac disease.


Diabetes | 2009

Majority of Children With Type 1 Diabetes Produce and Deposit Anti-Tissue Transglutaminase Antibodies in the Small Intestine

Mariantonia Maglio; Fiorella Florian; Monica Vecchiet; Renata Auricchio; Francesco Paparo; Raffaella Spadaro; Delia Zanzi; L. Rapacciuolo; Adriana Franzese; Daniele Sblattero; Roberto Marzari; Riccardo Troncone

OBJECTIVE Anti-tissue transglutaminase (TG2) antibodies are the serological marker of celiac disease. Given the close association between celiac disease and type 1 diabetes, we investigated the production and deposition of anti-TG2 antibodies in the jejunal mucosa of type 1 diabetic children. RESEARCH DESIGN AND METHODS Intestinal biopsies were performed in 33 type 1 diabetic patients with a normal mucosal architecture: 14 had high levels (potential celiac disease patients) and 19 had normal levels of serum anti-TG2 antibodies. All biopsy specimens were investigated for intestinal deposits of IgA anti-TG2 antibodies by double immunofluorescence. In addition, an antibody analysis using the phage display technique was performed on the intestinal biopsy specimens from seven type 1 diabetic patients, of whom four had elevated and three had normal levels of serum anti-TG2 antibodies. RESULTS Immunofluorescence studies showed that 11 of 14 type 1 diabetic children with elevated levels and 11 of 19 with normal serum levels of anti-TG2 antibodies presented with mucosal deposits of such autoantibodies. The phage display analysis technique confirmed the intestinal production of the anti-TG2 antibodies; however, whereas the serum-positive type 1 diabetic patients showed a preferential use of the VH5 antibody gene family, in the serum-negative patients the anti-TG2 antibodies belonged to the VH1 and VH3 families, with a preferential use of the latter. CONCLUSIONS Our findings demonstrate that there is intestinal production and deposition of anti-TG2 antibodies in the jejunal mucosa of the majority of type 1 diabetic patients. However, only those with elevated serum levels of anti-TG2 antibodies showed the VH usage that is typical of the anti-TG2 antibodies that are produced in patients with celiac disease.


The American Journal of Clinical Nutrition | 2013

An undigested gliadin peptide activates innate immunity and proliferative signaling in enterocytes: the role in celiac disease

Merlin Nanayakkara; Giuliana Lania; Mariantonia Maglio; Valentina Discepolo; M. Sarno; A. Gaito; Riccardo Troncone; Salvatore Auricchio; Renata Auricchio; Maria Vittoria Barone

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 others 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.


Scandinavian Journal of Gastroenterology | 2011

Immunogenicity of two oat varieties, in relation to their safety for celiac patients

Mariantonia Maglio; Giuseppe Mazzarella; Maria Vittoria Barone; Carmen Gianfrani; Norberto Pogna; Laura Gazza; Rosita Stefanile; Alessandra Camarca; B. Colicchio; Merlin Nanayakkara; Erasmo Miele; Gaetano Iaquinto; Nicola Giardullo; Francesco Maurano; Pasquale Santoro; Riccardo Troncone; Salvatore Auricchio

Abstract 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.

Collaboration


Dive into the Mariantonia Maglio's collaboration.

Top Co-Authors

Avatar

Riccardo Troncone

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Renata Auricchio

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Salvatore Auricchio

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

A. Tosco

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Erasmo Miele

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Francesco Paparo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Giuliana Lania

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Maria Vittoria Barone

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Merlin Nanayakkara

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Valentina Discepolo

University of Naples Federico II

View shared research outputs
Researchain Logo
Decentralizing Knowledge