Network


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

Hotspot


Dive into the research topics where Ruggiero Francavilla is active.

Publication


Featured researches published by Ruggiero Francavilla.


Nutrients | 2013

Non-Celiac Gluten Sensitivity: The New Frontier of Gluten Related Disorders

Carlo Catassi; Julio C. Bai; Bruno Bonaz; Gerd Bouma; Antonio Calabrò; Antonio Carroccio; Gemma Castillejo; Carolina Ciacci; Fernanda Cristofori; Jernej Dolinsek; Ruggiero Francavilla; Luca Elli; Peter H. Green; Wolfgang Holtmeier; Peter Koehler; Sibylle Koletzko; Christof Meinhold; David S. Sanders; Michael Schumann; Detlef Schuppan; Reiner Ullrich; Andreas Vécsei; Umberto Volta; Victor Zevallos; Anna Sapone; Alessio Fasano

Non Celiac Gluten sensitivity (NCGS) was originally described in the 1980s and recently a “re-discovered” disorder characterized by intestinal and extra-intestinal symptoms related to the ingestion of gluten-containing food, in subjects that are not affected with either celiac disease (CD) or wheat allergy (WA). Although NCGS frequency is still unclear, epidemiological data have been generated that can help establishing the magnitude of the problem. Clinical studies further defined the identity of NCGS and its implications in human disease. An overlap between the irritable bowel syndrome (IBS) and NCGS has been detected, requiring even more stringent diagnostic criteria. Several studies suggested a relationship between NCGS and neuropsychiatric disorders, particularly autism and schizophrenia. The first case reports of NCGS in children have been described. Lack of biomarkers is still a major limitation of clinical studies, making it difficult to differentiate NCGS from other gluten related disorders. Recent studies raised the possibility that, beside gluten, wheat amylase-trypsin inhibitors and low-fermentable, poorly-absorbed, short-chain carbohydrates can contribute to symptoms (at least those related to IBS) experienced by NCGS patients. In this paper we report the major advances and current trends on NCGS.


Journal of Pediatric Gastroenterology and Nutrition | 2010

Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives.

Laura de Magistris; V. Familiari; Antonio Pascotto; Anna Sapone; Alessandro Frolli; Patrizia Iardino; Maria Cartenì; Mario De Rosa; Ruggiero Francavilla; Gabriele Riegler; Roberto Militerni; Carmela Bravaccio

Objectives: Intestinal permeability (IPT) was investigated in patients with autism as well as in their first-degree relatives to investigate leaky gut hypothesis. Faecal calprotectin (FC) was also measured in patients with autism, either with or without gastrointestinal symptoms, and in their first-degree relatives. Patients and Methods: IPT results, assessed by means of the lactulose/mannitol test, were compared with adult and child controls and with FC values. Results: A high percentage of abnormal IPT values were found among patients with autism (36.7%) and their relatives (21.2%) compared with normal subjects (4.8%). Patients with autism on a reported gluten-casein–free diet had significantly lower IPT values compared with those who were on an unrestricted diet and controls. Gastrointestinal symptoms were present in 46.7% of children with autism: constipation (45.5%), diarrhoea (34.1%), and others (alternating diarrhoea/constipation, abdominal pain, etc: 15.9%). FC was elevated in 24.4% of patients with autism and in 11.6% of their relatives; it was not, however, correlated with abnormal IPT values. Conclusions: The results obtained support the leaky gut hypothesis and indicate that measuring IPT could help to identify a subgroup of patients with autism who could benefit from a gluten-free diet. The IPT alterations found in first-degree relatives suggest the presence of an intestinal (tight-junction linked) hereditary factor in the families of subjects with autism.


The New England Journal of Medicine | 2014

Introduction of Gluten, HLA Status, and the Risk of Celiac Disease in Children

Abstr Act; Elena Lionetti; Stefania Castellaneta; Ruggiero Francavilla; Alfredo Pulvirenti; Elio Tonutti; Sergio Amarri; Maria Barbato; Cristiana Barbera; Graziano Barera; Antonella Bellantoni; Emanuela Castellano; Graziella Guariso; Maria Giovanna Limongelli; Salvatore Pellegrino; Carlo Polloni; Claudio Ughi; Giovanna Zuin; Alessio Fasano; Carlo Catassi

BACKGROUND The relationship between the risk of celiac disease and both the age at which gluten is introduced to a childs diet and a childs early dietary pattern is unclear. METHODS We randomly assigned 832 newborns who had a first-degree relative with celiac disease to the introduction of dietary gluten at 6 months (group A) or 12 months (group B). The HLA genotype was determined at 15 months of age, and serologic screening for celiac disease was evaluated at 15, 24, and 36 months and at 5, 8, and 10 years. Patients with positive serologic findings underwent intestinal biopsies. The primary outcome was the prevalence of celiac disease autoimmunity and of overt celiac disease among the children at 5 years of age. RESULTS Of the 707 participants who remained in the trial at 36 months, 553 had a standard-risk or high-risk HLA genotype and completed the study. At 2 years of age, significantly higher proportions of children in group A than in group B had celiac disease autoimmunity (16% vs. 7%, P=0.002) and overt celiac disease (12% vs. 5%, P=0.01). At 5 years of age, the between-group differences were no longer significant for autoimmunity (21% in group A and 20% in group B, P=0.59) or overt disease (16% and 16%, P=0.78 by the log-rank test). At 10 years, the risk of celiac disease autoimmunity was far higher among children with high-risk HLA than among those with standard-risk HLA (38% vs. 19%, P=0.001), as was the risk of overt celiac disease (26% vs. 16%, P=0.05). Other variables, including breast-feeding, were not associated with the development of celiac disease. CONCLUSIONS Neither the delayed introduction of gluten nor breast-feeding modified the risk of celiac disease among at-risk infants, although the later introduction of gluten was associated with a delayed onset of disease. A high-risk HLA genotype was an important predictor of disease. (Funded by the Fondazione Celiachia of the Italian Society for Celiac Disease; CELIPREV ClinicalTrials.gov number, NCT00639444.).


PLOS ONE | 2013

Fecal Microbiota and Metabolome of Children with Autism and Pervasive Developmental Disorder Not Otherwise Specified

Maria De Angelis; Maria Teresa Piccolo; Lucia Vannini; Sonya Siragusa; Andrea De Giacomo; Diana Isabella Serrazzanetti; Fernanda Cristofori; Maria Elisabetta Guerzoni; Marco Gobbetti; Ruggiero Francavilla

This study aimed at investigating the fecal microbiota and metabolome of children with Pervasive Developmental Disorder Not Otherwise Specified (PDD-NOS) and autism (AD) in comparison to healthy children (HC). Bacterial tag-encoded FLX-titanium amplicon pyrosequencing (bTEFAP) of the 16S rDNA and 16S rRNA analyses were carried out to determine total bacteria (16S rDNA) and metabolically active bacteria (16S rRNA), respectively. The main bacterial phyla (Firmicutes, Bacteroidetes, Fusobacteria and Verrucomicrobia) significantly (P<0.05) changed among the three groups of children. As estimated by rarefaction, Chao and Shannon diversity index, the highest microbial diversity was found in AD children. Based on 16S-rRNA and culture-dependent data, Faecalibacterium and Ruminococcus were present at the highest level in fecal samples of PDD-NOS and HC children. Caloramator, Sarcina and Clostridium genera were the highest in AD children. Compared to HC, the composition of Lachnospiraceae family also differed in PDD-NOS and, especially, AD children. Except for Eubacterium siraeum, the lowest level of Eubacteriaceae was found on fecal samples of AD children. The level of Bacteroidetes genera and some Alistipes and Akkermansia species were almost the highest in PDD-NOS or AD children as well as almost all the identified Sutterellaceae and Enterobacteriaceae were the highest in AD. Compared to HC children, Bifidobacterium species decreased in AD. As shown by Canonical Discriminant Analysis of Principal Coordinates, the levels of free amino acids and volatile organic compounds of fecal samples were markedly affected in PDD-NOS and, especially, AD children. If the gut microbiota differences among AD and PDD-NOS and HC children are one of the concomitant causes or the consequence of autism, they may have implications regarding specific diagnostic test, and/or for treatment and prevention.


The Journal of Pediatrics | 2008

The effects of probiotics on feeding tolerance, bowel habits, and gastrointestinal motility in preterm newborns.

Flavia Indrio; Giuseppe Riezzo; Francesco Raimondi; Massimo Bisceglia; Luciano Cavallo; Ruggiero Francavilla

OBJECTIVE To investigate the effect of dietary supplementation with a probiotic on feeding tolerance and gastrointestinal motility in healthy formula-fed preterm infants. STUDY DESIGN Thirty preterm newborns were enrolled; 10 were exclusively breast-fed, and the remaining 20 were randomly assigned in a double-blind manner to receive either Lactobacillus reuteri ATCC 55730 (at dose of 1 x 10(8) colony forming units a day) or placebo for 30 days. Clinical symptoms of gastrointestinal function (regurgitation, vomiting, inconsolable crying, and evacuation) and physiological variables (gastric electrical activity and emptying) were recorded before and after the dietary intervention. RESULTS Body weight gains per day were similar for the 3 groups, and no adverse events were recorded. Newborns receiving probiotics showed a significant decrease in regurgitation and mean daily crying time and a larger number of stools compared with those given placebo. Gastric emptying rate was significantly increased, and fasting antral area was significantly reduced in both the newborns receiving L. reuteri and breast-fed newborns compared with placebo. CONCLUSIONS Our results suggest a useful role for L. reuteri supplementation in improving feeding tolerance and gut function in formula-fed preterm newborns.


BMC Microbiology | 2011

Duodenal and faecal microbiota of celiac children: molecular, phenotype and metabolome characterization

Raffaella Di Cagno; Maria De Angelis; Ilaria De Pasquale; Maurice Ndagijimana; Pamela Vernocchi; Patrizia Ricciuti; F. Gagliardi; Luca Laghi; Carmine Crecchio; Maria Elisabetta Guerzoni; Marco Gobbetti; Ruggiero Francavilla

BackgroundEpidemiology of celiac disease (CD) is increasing. CD mainly presents in early childhood with small intestinal villous atrophy and signs of malabsorption. Compared to healthy individuals, CD patients seemed to be characterized by higher numbers of Gram-negative bacteria and lower numbers Gram-positive bacteria.ResultsThis study aimed at investigating the microbiota and metabolome of 19 celiac disease children under gluten-free diet (treated celiac disease, T-CD) and 15 non-celiac children (HC). PCR-denaturing gradient gel electrophoresis (DGGE) analyses by universal and group-specific primers were carried out in duodenal biopsies and faecal samples. Based on the number of PCR-DGGE bands, the diversity of Eubacteria was the higher in duodenal biopsies of T-CD than HC children. Bifidobacteria were only found in faecal samples. With a few exceptions, PCR-DGGE profiles of faecal samples for Lactobacillus and Bifidobacteria differed between T-CD and HC. As shown by culture-dependent methods, the levels of Lactobacillus, Enterococcus and Bifidobacteria were confirmed to be significantly higher (P = 0.028; P = 0.019; and P = 0.023, respectively) in fecal samples of HC than in T-CD children. On the contrary, cell counts (CFU/ml) of presumptive Bacteroides, Staphylococcus, Salmonella, Shighella and Klebsiella were significantly higher (P = 0.014) in T-CD compared to HC children. Enterococcus faecium and Lactobacillus plantarum were the species most diffusely identified. This latter species was also found in all duodenal biopsies of T-CD and HC children. Other bacterial species were identified only in T-CD or HC faecal samples. As shown by Randomly Amplified Polymorphic DNA-PCR analysis, the percentage of strains identified as lactobacilli significantly (P = 0.011) differed between T-CD (ca. 26.5%) and HC (ca. 34.6%) groups. The metabolome of T-CD and HC children was studied using faecal and urine samples which were analyzed by gas-chromatography mass spectrometry-solid-phase microextraction and 1H-Nuclear Magnetic Resonance. As shown by Canonical Discriminant Analysis of Principal Coordinates, the levels of volatile organic compounds and free amino acids in faecal and/or urine samples were markedly affected by CD.ConclusionAs shown by the parallel microbiology and metabolome approach, the gluten-free diet lasting at least two years did not completely restore the microbiota and, consequently, the metabolome of CD children. Some molecules (e.g., ethyl-acetate and octyl-acetate, some short chain fatty acids and free amino acids, and glutamine) seems to be metabolic signatures of CD.


Nutrients | 2015

Diagnosis of Non-Celiac Gluten Sensitivity (NCGS): The Salerno Experts' Criteria.

Carlo Catassi; Luca Elli; Bruno Bonaz; Gerd Bouma; Antonio Carroccio; Gemma Castillejo; Christophe Cellier; Fernanda Cristofori; Laura de Magistris; Jernej Dolinsek; Walburga Dieterich; Ruggiero Francavilla; Marios Hadjivassiliou; Wolfgang Holtmeier; Ute Körner; Daniel A. Leffler; Knut E.A. Lundin; Giuseppe Mazzarella; Chris Jj Mulder; Nicoletta Pellegrini; Kamran Rostami; David S. Sanders; Gry I. Skodje; Detlef Schuppan; Reiner Ullrich; Umberto Volta; Marianne Williams; Victor Zevallos; Yurdagül Zopf; Alessio Fasano

Non-Celiac Gluten Sensitivity (NCGS) is a syndrome characterized by intestinal and extra-intestinal symptoms related to the ingestion of gluten-containing food, in subjects that are not affected by either celiac disease or wheat allergy. Given the lack of a NCGS biomarker, there is the need for standardizing the procedure leading to the diagnosis confirmation. In this paper we report experts’ recommendations on how the diagnostic protocol should be performed for the confirmation of NCGS. A full diagnostic procedure should assess the clinical response to the gluten-free diet (GFD) and measure the effect of a gluten challenge after a period of treatment with the GFD. The clinical evaluation is performed using a self-administered instrument incorporating a modified version of the Gastrointestinal Symptom Rating Scale. The patient identifies one to three main symptoms that are quantitatively assessed using a Numerical Rating Scale with a score ranging from 1 to 10. The double-blind placebo-controlled gluten challenge (8 g/day) includes a one-week challenge followed by a one-week washout of strict GFD and by the crossover to the second one-week challenge. The vehicle should contain cooked, homogeneously distributed gluten. At least a variation of 30% of one to three main symptoms between the gluten and the placebo challenge should be detected to discriminate a positive from a negative result. The guidelines provided in this paper will help the clinician to reach a firm and positive diagnosis of NCGS and facilitate the comparisons of different studies, if adopted internationally.


Pediatrics | 2010

A Randomized Controlled Trial of Lactobacillus GG in Children With Functional Abdominal Pain

Ruggiero Francavilla; Miniello; A.M. Magistà; De Canio A; N. Bucci; Gagliardi F; Elena Lionetti; Stefania Castellaneta; Lorenzo Polimeno; Peccarisi L; Flavia Indrio; Luciano Cavallo

OBJECTIVE: Our aim was to determine whether Lactobacillus rhamnosus GG (LGG) relieves symptoms in children with recurrent abdominal pain. PATIENTS AND METHODS: A total of 141 children with irritable bowel syndrome (IBS) or functional pain were enrolled in 9 primary care sites and a referral center. Children entered a randomized, double-blind, placebo-controlled trial and received LGG or placebo for 8 weeks and entered follow-up for 8 weeks. The primary outcome was overall pain at the end of the intervention period. At entry and at the end of the trial, children underwent a double-sugar intestinal permeability test. RESULTS: Compared with baseline, LGG, but not placebo, caused a significant reduction of both frequency (P < .01) and severity (P < .01) of abdominal pain. These differences still were significant at the end of follow-up (P < .02 and P < .001, respectively). At week 12, treatment success was achieved in 48 children in the LGG group compared with 37 children in the placebo group (P < .03); this difference still was present at the end of follow-up (P < .03). At entry, 59% of the children had abnormal results from the intestinal permeability test; LGG, but not placebo, determined a significant decrease in the number of patients with abnormal results from the intestinal permeability testing (P < .03). These effects mainly were in children with IBS. CONCLUSIONS: LGG significantly reduces the frequency and severity of abdominal pain in children with IBS; this effect is sustained and may be secondary to improvement of the gut barrier.


Alimentary Pharmacology & Therapeutics | 2006

Lactobacillus reuteri therapy to reduce side-effects during anti-Helicobacter pylori treatment in children: a randomized placebo controlled trial.

Elena Lionetti; Vito Leonardo Miniello; Stefania Castellaneta; A.M. Magistà; A. De Canio; Giovanni Maurogiovanni; Enzo Ierardi; Luciano Cavallo; Ruggiero Francavilla

Helicobacter pylori eradication fails in about 25–30% of children, particularly because of the occurrence of resistance to antibiotics and side‐effects.


Italian Journal of Pediatrics | 2013

Gastrointestinal function development and microbiota.

Antonio Di Mauro; Josef Neu; Giuseppe Riezzo; Francesco Raimondi; Domenico Martinelli; Ruggiero Francavilla; Flavia Indrio

The intestinal microbiota plays an important role in the development of post-natal gastrointestinal functions of the host. Recent advances in our capability to identify microbes and their function offer exciting opportunities to evaluate the complex cross talk between microbiota, intestinal barrier, immune system and the gut-brain axis. This review summarizes these interactions in the early colonization of gastrointestinal tract with a major focus on the role of intestinal microbiota in the pathogenesis of feeding intolerance in preterm newborn. The potential benefit of early probiotic supplementation opens new perspectives in case of altered intestinal colonization at birth as preventive and therapeutic agents.

Collaboration


Dive into the Ruggiero Francavilla's collaboration.

Researchain Logo
Decentralizing Knowledge