Network


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

Hotspot


Dive into the research topics where Franco Scaldaferri is active.

Publication


Featured researches published by Franco Scaldaferri.


Gastroenterology | 2009

VEGF-A links angiogenesis and inflammation in inflammatory bowel disease pathogenesis.

Franco Scaldaferri; Stefania Vetrano; Miquel Sans; Vincenzo Arena; Giuseppe Straface; Egidio Stigliano; Alessandro Repici; Andreas Sturm; Alberto Malesci; Julián Panés; Seppo Yla–Herttuala; Claudio Fiocchi; Silvio Danese

BACKGROUND & AIMS Vascular endothelial growth factor A (VEGF-A) mediates angiogenesis and might also have a role in inflammation and immunity. We examined whether VEGF-A signaling has a role in inflammatory bowel disease (IBD). METHODS Expression levels of VEGF-A, and its receptors VEGFR-1 and VEGFR-2, were examined in samples from patients with IBD and compared with those of controls. The capacity of VEGF-A to induce angiogenesis was tested in human intestinal microvascular endothelial cells using cell-migration and matrigel tubule-formation assays. Levels of vascular cellular adhesion molecule-1 and intercellular adhesion molecule were measured by flow cytometry to determine induction of inflammation; neutrophil adhesion was also assayed. Expression patterns were determined in tissues from mice with dextran sulfate sodium (DSS)-induced colitis; the effects of VEGF-A overexpression and blockade were assessed in these mice by adenoviral transfer of VEGF-A and soluble VEGFR-1. Intestinal angiogenesis was measured by quantitative CD31 staining and leukocyte adhesion in vivo by intravital microscopy. RESULTS Levels of VEGF-A and VEGFR-2 increased in samples from patients with IBD and colitic mice. VEGF-A induced angiogenesis of human intestinal microvascular endothelial cells in vitro as well as an inflammatory phenotype and adherence of neutrophils to intestinal endothelium. Overexpression of VEGF-A in mice with DSS-induced colitis worsened their condition, whereas overexpression of soluble VEGFR-1 had the opposite effect. Furthermore, overexpression of VEGF-A increased mucosal angiogenesis and stimulated leukocyte adhesion in vivo. CONCLUSIONS VEGF-A appears to be a novel mediator of IBD by promoting intestinal angiogenesis and inflammation. Agents that block VEGF-A signaling might reduce intestinal inflammation in patients with IBD.


Gut Pathogens | 2013

Commensal Clostridia: leading players in the maintenance of gut homeostasis

Loris Riccardo Lopetuso; Franco Scaldaferri; Valentina Petito; Antonio Gasbarrini

The gastrointestinal tract is a complex and dynamic network where an intricate and mutualistic symbiosis modulates the relationship between the host and the microbiota in order to establish and ensure gut homeostasis. Commensal Clostridia consist of gram-positive, rod-shaped bacteria in the phylum Firmicutes and make up a substantial part of the total bacteria in the gut microbiota. They start to colonize the intestine of breastfed infants during the first month of life and populate a specific region in the intestinal mucosa in close relationship with intestinal cells. This position allows them to participate as crucial factors in modulating physiologic, metabolic and immune processes in the gut during the entire lifespan, by interacting with the other resident microbe populations, but also by providing specific and essential functions. This review focus on what is currently known regarding the role of commensal Clostridia in the maintenance of overall gut function, as well as touch on their potential contribution in the unfavorable alteration of microbiota composition (dysbiosis) that has been implicated in several gastrointestinal disorders. Commensal Clostridia are strongly involved in the maintenance of overall gut function. This leads to important translational implications in regard to the prevention and treatment of dysbiosis, to drug efficacy and toxicity, and to the development of therapies that may modulate the composition of the microflora, capitalizing on the key role of commensal Clostridia, with the end goal of promoting gut health.


Journal of Immunology | 2006

TNF-α Blockade Down-Regulates the CD40/CD40L Pathway in the Mucosal Microcirculation: A Novel Anti-Inflammatory Mechanism of Infliximab in Crohn’s Disease

Silvio Danese; Miquel Sans; Franco Scaldaferri; Alessandro Sgambato; Sergio Rutella; Achille Cittadini; Josep M. Piqué; Julián Panés; Jeffry A. Katz; Antonio Gasbarrini; Claudio Fiocchi

The CD40/CD40 ligand (CD40L) pathway is involved in Crohn’s disease (CD) pathogenesis. In the patients’ circulation, soluble CD40L (sCD40L) levels are elevated and surface CD40L is increased in platelets and T cells, whereas in the intestine CD40 is overexpressed in the microvasculature and CD40L in platelets and T cells. The therapeutic effects of infliximab in CD are attributed to its systemic anti-TNF-α action, but because TNF-α modulates both CD40 and CD40L, we investigated whether infliximab affects the CD40/CD40L pathway in the intestine. Eighteen CD patients were evaluated before and after infliximab therapy. Plasma sCD40L was measured by ELISA and platelet and peripheral blood T cell (PBT) CD40L expression by flow cytometry. Microvascular CD40 and VCAM-1 expression were assessed in mucosal biopsies by immunohistochemistry and by flow cytometry in human intestinal microvascular endothelial cells (HIMEC). Cell cultures were performed in the presence and absence of infliximab. Infliximab treatment significantly reduced plasma sCD40L levels and eliminated CD40 and VCAM-1 from mucosal microvessels. In vitro infliximab prevented TNF-α-induced CD40 and VCAM-1 expression by HIMEC, and reduced PBT, but not platelet, surface CD40L expression and sCD40L release. In addition, infliximab decreased T cell-induced VCAM-1 expression in HIMEC by down-regulating CD40L in T cells and promoting T cells apoptosis. These findings point to a novel mechanism of action of infliximab, i.e., the disruption of CD40/CD40L-dependent cognate interactions between intestinal microvessels and T cells. Thus, in addition to neutralizing TNF-α and inducing T cell death, the therapeutic effects of infliximab in CD appear to be also mediated by inhibition of vascular inflammation in the gut.


BioMed Research International | 2013

Gut Microbial Flora, Prebiotics, and Probiotics in IBD: Their Current Usage and Utility

Franco Scaldaferri; Viviana Gerardi; Loris Riccardo Lopetuso; Fabio Del Zompo; Francesca Mangiola; Ivo Boskoski; Giovanni Bruno; Valentina Petito; Lucrezia Laterza; Giovanni Cammarota; Eleonora Gaetani; Alessandro Sgambato; Antonio Gasbarrini

Inflammatory bowel diseases are chronic diseases affecting the gastrointestinal tract, whose major forms are represented by Crohns disease (CD) and ulcerative colitis (UC). Their etiology is still unclear, although several factors have been identified as major determinants for induction or relapses. Among these, the role of the “forgotten organ”, gut microbiota, has become more appreciated in recent years. The delicate symbiotic relationship between the gut microbiota and the host appears to be lost in IBD. In this perspective, several studies have been conducted to assess the role of prebiotics and probiotics in gut microbiota modulation. This is a minireview aimed to address in an easy format (simple questions-simple answers) some common issues about the theme. An update on the role of selected constituents of gut microbiota in the pathogenesis of IBD is presented together with the analysis of the efficacy of gut microbiota modulation by prebiotics and probiotics administration in the management of IBD.


Gut | 2017

European consensus conference on faecal microbiota transplantation in clinical practice.

Giovanni Cammarota; Gianluca Ianiro; Herbert Tilg; Mirjana Rajilić-Stojanović; Patrizia Kump; Reetta Satokari; Harry Sokol; Perttu Arkkila; Cristina Pintus; Ailsa Hart; Jonathan Segal; Marina Aloi; Luca Masucci; A. Molinaro; Franco Scaldaferri; Giovanni Gasbarrini; Antonio Lopez-Sanroman; Alexander Link; Pieter F. de Groot; Willem M. de Vos; Christoph Högenauer; Peter Malfertheiner; Eero Mattila; Tomica Milosavljevic; Max Nieuwdorp; Maurizio Sanguinetti; Magnus Simren; Antonio Gasbarrini

Faecal microbiota transplantation (FMT) is an important therapeutic option for Clostridium difficile infection. Promising findings suggest that FMT may play a role also in the management of other disorders associated with the alteration of gut microbiota. Although the health community is assessing FMT with renewed interest and patients are becoming more aware, there are technical and logistical issues in establishing such a non-standardised treatment into the clinical practice with safety and proper governance. In view of this, an evidence-based recommendation is needed to drive the practical implementation of FMT. In this European Consensus Conference, 28 experts from 10 countries collaborated, in separate working groups and through an evidence-based process, to provide statements on the following key issues: FMT indications; donor selection; preparation of faecal material; clinical management and faecal delivery and basic requirements for implementing an FMT centre. Statements developed by each working group were evaluated and voted by all members, first through an electronic Delphi process, and then in a plenary consensus conference. The recommendations were released according to best available evidence, in order to act as guidance for physicians who plan to implement FMT, aiming at supporting the broad availability of the procedure, discussing other issues relevant to FMT and promoting future clinical research in the area of gut microbiota manipulation. This consensus report strongly recommends the implementation of FMT centres for the treatment of C. difficile infection as well as traces the guidelines of technicality, regulatory, administrative and laboratory requirements.


Journal of Clinical Investigation | 2007

Crucial role of the protein C pathway in governing microvascular inflammation in inflammatory bowel disease

Franco Scaldaferri; Miquel Sans; Stefania Vetrano; C. Graziani; Raimondo De Cristofaro; Bruce Gerlitz; Alessandro Repici; Vincenzo Arena; Alberto Malesci; Julián Panés; Brian W. Grinnell; Silvio Danese

Endothelial protein C receptor (EPCR) and thrombomodulin (TM) are expressed at high levels in the resting microvasculature and convert protein C (PC) into its activated form, which is a potent anticoagulant and antiinflammatory molecule. Here we provide evidence that in Crohn disease (CD) and ulcerative colitis (UC), the 2 major forms of inflammatory bowel disease (IBD), there was loss of expression of endothelial EPCR and TM, which in turns caused impairment of PC activation by the inflamed mucosal microvasculature. In isolated human intestinal endothelial cells, administration of recombinant activated PC had a potent antiinflammatory effect, as demonstrated by downregulated cytokine-dependent cell adhesion molecule expression and chemokine production as well as inhibited leukocyte adhesion. In vivo, administration of activated PC was therapeutically effective in ameliorating experimental colitis as evidenced by reduced weight loss, disease activity index, and histological colitis scores as well as inhibited leukocyte adhesion to the inflamed intestinal vessels. The results suggest that the PC pathway represents a new system crucially involved in governing intestinal homeostasis mediated by the mucosal microvasculature. Restoring the PC pathway may represent a new therapeutic approach to suppress intestinal inflammation in IBD.


The American Journal of Gastroenterology | 2005

Homocysteine triggers mucosal microvascular activation in inflammatory bowel disease

Silvio Danese; Alessandro Sgambato; Alfredo Papa; Franco Scaldaferri; Roberto Pola; Miquel Sans; Maria Lovecchio; Giovanni Gasbarrini; Achille Cittadini; Antonio Gasbarrini

OBJECTIVES:Increased homocysteine contributes to the pathophysiology of several chronic inflammatory diseases. Whether homocysteine could participate in mucosal inflammation in inflammatory bowel disease (IBD) has not been explored yet. Our aims were to study the levels of plasma and mucosal homocysteine in IBD patients and to assess whether homocysteine can trigger an inflammatory reaction on human intestinal microvascular endothelial cells (HIMECs).METHODS:Homocysteine was measured in the plasma, mucosal biopsy, and lamina propria mononuclear cell (LPMC) supernatants from normal and IBD subjects. HIMEC were cultured in presence of homocysteine, TNF-α, or folic acid, alone or in combination. Expression of vascular cell adhesion molecule 1 (VCAM-1) and intercellular cell adhesion molecule 1 was measured by flow cytometry and monocyte chemoattractant protein-1 (MCP-1) production by ELISA. Phosphorylation of p38 and p42/44 was assessed by immunoblot in HIMEC extracts. T-cell- and monocyte-HIMEC adhesion assays were used to evaluate the impact of homocysteine on leukocyte adhesion to intestinal endothelial cells.RESULTS:Patients with IBD displayed significantly higher homocysteine plasma and mucosal levels than control subjects. IBD-derived LPMC released higher homocysteine than control-derived LPMC. Treatment of HIMEC with homocysteine, and synergistically with the combination of TNF-α and homocysteine, triggered HIMEC inflammation, resulting in VCAM-1 up-regulation, MCP-1 production, and p38 phosphorylation. These events lead to an increased capacity of HIMEC to adhere T- and monocyte cells and were blocked by folic acid treatment.CONCLUSIONS:Homocysteine is increased in both the mucosa and plasma of patients with Crohns disease and ulcerative colitis and contributes to the inflammatory state of the mucosal IBD endothelium. Therefore, homocysteine could play a proinflammatory role in IBD, which can be efficiently targeted by folic acid supplementation.


Journal of Crohns & Colitis | 2015

European Evidence-based Consensus: Inflammatory Bowel Disease and Malignancies

Vito Annese; Laurent Beaugerie; Laurence J. Egan; L. Biancone; Claus Bolling; Christian Brandts; Daan Dierickx; Reinhard Dummer; Gionata Fiorino; Jean–Marc Gornet; Peter D. Higgins; Konstantinos Katsanos; Loes Nissen; Gianluca Pellino; Gerhard Rogler; Franco Scaldaferri; Edyta Szymanska; Rami Eliakim

The global prevalence of cancer is increasing, largely as more patients are living into old age. Therefore, gastroenterologists caring for patients with inflammatory bowel disease [IBD] increasingly are managing patients with cancer, or a previous history of cancer. This often requires joint management with the patient’s oncologist, enabling case-by-case decision-making based on the characteristics and expected evolution of the index cancer. Previously, no European guidelines existed describing the impact of IBD on malignancy. For this reason, the European Crohn’s and Colitis Organisation [ECCO] Guidelines Committee [GuiCom] decided to elaborate a set of Consensus Statements on optimal risk/benefit strategies for treating IBD patients with cancer or a history of cancer. The development of clinical practice guidelines is expensive and time consuming, and it is the Committee’s hope that these statements will facilitate and accelerate future efforts to elaborate formal guidelines, providing useful information on areas for which evidence is lacking and where controlled studies are needed. The strategy used to produce the Consensus Statements involved five steps: 1. Two members of Guicom [VA and RE] identified four main topics that needed to be addressed: a] IBD and solid tumours; b] IBD and skin and haematological malignancies; c] malignancy related to therapy: risk and prevention; and d] management of IBD patients with a history of malignancy. During 2014, calls for participation in the drafting of consensus statements were issued to ECCO members, and selected oncologists known for their expertise and active research in the field were invited to join the Consensus. Participants were selected by the Committee, and four working groups were created, each composed of a chairperson [LE, RE, LB, and VA], two ECCO members including young members [Y-ECCO], and one or two experienced oncologists. The chairmen and their groups elaborated relevant questions on topics dealing with current practice and/or areas …


Journal of Clinical Gastroenterology | 2012

The gut barrier: new acquisitions and therapeutic approaches.

Franco Scaldaferri; Marco Pizzoferrato; Gerardi; Loris Riccardo Lopetuso; Antonio Gasbarrini

The intestinal barrier serves 2 critical functions for the survival of the individual: first, it allows nutrient absorption and second, it defends the body from dangerous macromolecule penetration. It is a complex multilayer system, consisting of an external “anatomic” barrier and an inner “functional” immunological barrier. The interaction of these 2 barriers enables equilibrated permeability to be maintained. Many factors can alter this balance: gut microflora modifications, mucus layer alterations, and epithelial damage can increase intestinal permeability, allowing the translocation of luminal content to the inner layer of intestinal wall. Several techniques are now available that enable us to study gut permeability: “in vitro” models (Caco-2 and HT29-MTX cells) and “in vivo” not invasive tests (sugar tests and radioisotope scanning tests) are used to estimate permeability and to suggest molecular pathophysiological mechanisms of intestinal permeability in health and diseases. Many medicinal products used in the treatment of gastrointestinal diseases have also found to play an active role in modulate intestinal permeability: corticosteroids, 5-aminosalicylic acid, anti–tumor necrosis factor, probiotics, and mucosal protectors, like gelatin tannate. This review will particularly address the role of the gut barrier in maintaining intestinal permeability (microbiota, mucus, and epithelial cells), the techniques used for estimating intestinal permeability and the therapeutic approaches able to modify it.


Gut | 2007

Critical role of the CD40–CD40-ligand pathway in regulating mucosal inflammation-driven angiogenesis in inflammatory bowel disease

Silvio Danese; Franco Scaldaferri; Stefania Vetrano; Tommaso Stefanelli; C. Graziani; Alessandro Repici; Riccardo Ricci; Giuseppe Straface; Alessandro Sgambato; Alberto Malesci; Claudio Fiocchi; Sergio Rutella

Background and aims: Angiogenesis is a novel component in inflammatory bowel disease (IBD) pathogenesis. We have previously shown that immune–nonimmune interactions through the CD40–CD40-ligand (CD40L) pathway might sustain gut inflammation, although their effect on regulating inflammation-driven angiogenesis is unknown. The present study evaluated the role of the CD40–CD40L interaction in the promotion of immune-mediated angiogenesis in IBD. Methods: Human nonimmune cells of colonic origin—namely, human intestinal fibroblasts (HIFs) and human intestinal microvascular endothelial cells (HIMECs)—were activated with either soluble CD40L (sCD40L), or CD40+ D1.1 cells or CD40L-activated lamina propria T (LPT) cells before measuring pro-angiogenic cytokine release. Blocking antibodies to either CD40 or CD40L were used to disrupt the CD40–CD40L interaction. The dextran sodium sulphate (DSS) model of experimental colitis in CD40 and CD40L knockout mice was established to assess whether the CD40–CD40L pathway was implicated in controlling inflammation-driven angiogenesis in vivo. Results: Engagement of CD40 on HIFs promoted the release of vascular endothelial growth factor (VEGF), interleukin-8 (IL-8) and hepatocyte growth factor (HGF). LPT cells were potent inducers of pro-angiogenic cytokine secretion by HIFs. Supernatants from sCD40L-activated HIFs induced migration of HIMECs and tubule formation, both of which were inhibited by blocking antibodies to either VEGF, IL-8 or HGF. Both CD40- and CD40L-deficient mice were protected from DSS-induced colitis and displayed a significant impairment of gut inflammation-driven angiogenesis, as assessed by microvascular density. Conclusions: The CD40–CD40L pathway appears to be crucially involved in regulating inflammation-driven angiogenesis, suggesting that strategies aimed at blocking CD40–CD40L interactions might be beneficial in acute and chronic intestinal injury.

Collaboration


Dive into the Franco Scaldaferri's collaboration.

Top Co-Authors

Avatar

Antonio Gasbarrini

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Loris Riccardo Lopetuso

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Valentina Petito

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giovanni Cammarota

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Alessandro Sgambato

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Viviana Gerardi

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Alfredo Papa

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Andrea Poscia

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

C. Graziani

Catholic University of the Sacred Heart

View shared research outputs
Researchain Logo
Decentralizing Knowledge