Network


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

Hotspot


Dive into the research topics where Daniele Veggi is active.

Publication


Featured researches published by Daniele Veggi.


Science Translational Medicine | 2011

Rational Design of a Meningococcal Antigen Inducing Broad Protective Immunity

Maria Scarselli; Beatrice Aricò; Brunella Brunelli; Silvana Savino; F. Di Marcello; E. Palumbo; Daniele Veggi; Laura Ciucchi; Elena Cartocci; Matthew J. Bottomley; Enrico Malito; P. Lo Surdo; Maurizio Chiron Srl Comanducci; Marzia Monica Giuliani; Francesca Cantini; Sara Dragonetti; A. Colaprico; F. Doro; P. Giannetti; Michele Pallaoro; B. Brogioni; Marta Tontini; M. Hilleringmann; Vincenzo Nardi-Dei; Lucia Banci; M Pizza; Rino Rappuoli

A single chimeric protein induces protective immunity against all meningococcal B strains with implications for producing broadly protective vaccines. All for One and One for All The three musketeers were a formidable team, but imagine combining all of their skills and valor into just one musketeer. That is precisely the approach that Rappuoli and his colleagues have taken with their design of a vaccine against meningococcus B, the bacterial pathogen that causes meningitis. Although mining of the genome sequence of this pathogen has yielded excellent targets that could be used in a vaccine, many of these antigens show a high degree of variation that has stymied attempts to use them as vaccine immunogens. For example, factor H binding protein is essential for the survival of meningococcus B in the human host because it protects the pathogen from the onslaught of the human immune system’s complement pathway. Because it is essential for survival, factor H binding protein should be a valuable immunogen, but because it has at least 300 sequence variants, it is impractical to make one vaccine that contains all of these variants. Rappuoli and his colleagues have tackled this problem by dividing the 300 sequence variants of factor H binding protein into three major groups. Using detailed structural information about these three major variants, they engineered variant 1 to carry patches of amino acids from the surfaces of variants 2 and 3. They then introduced groups of point mutations into the amino acids of these transplanted patches to mimic the natural variation of variant 2 and 3 strains of meningococcus B. They then tested which of the 54 engineered single chimeric immunogens could elicit bactericidal antibodies against many different strains of meningococcus B. To do this, they injected the immunogens into mice and assayed mouse sera in vitro for bactericidal activity against multiple bacterial strains. One chimeric immunogen, called G1, was capable of inducing bactericidal antibodies that could kill all strains of meningococcus B, suggesting that it could be used to produce a broadly protective vaccine. This structure-based approach to vaccine design may be useful not only for meningococcus B but also for other pathogens like HIV that show a high degree of antigenic variation. The sequence variability of protective antigens is a major challenge to the development of vaccines. For Neisseria meningitidis, the bacterial pathogen that causes meningitis, the amino acid sequence of the protective antigen factor H binding protein (fHBP) has more than 300 variations. These sequence differences can be classified into three distinct groups of antigenic variants that do not induce cross-protective immunity. Our goal was to generate a single antigen that would induce immunity against all known sequence variants of N. meningitidis. To achieve this, we rationally designed, expressed, and purified 54 different mutants of fHBP and tested them in mice for the induction of protective immunity. We identified and determined the crystal structure of a lead chimeric antigen that was able to induce high levels of cross-protective antibodies in mice against all variant strains tested. The new fHBP antigen had a conserved backbone that carried an engineered surface containing specificities for all three variant groups. We demonstrate that the structure-based design of multiple immunodominant antigenic surfaces on a single protein scaffold is possible and represents an effective way to create broadly protective vaccines.


Infection and Immunity | 2009

Factor H-Binding Protein Is Important for Meningococcal Survival in Human Whole Blood and Serum and in the Presence of the Antimicrobial Peptide LL-37

Kate L. Seib; Davide Serruto; Francesca Oriente; Isabel Delany; Jeannette Adu-Bobie; Daniele Veggi; Beatrice Aricò; Rino Rappuoli; Mariagrazia Pizza

ABSTRACT Factor H-binding protein (fHBP; GNA1870) is one of the antigens of the recombinant vaccine against serogroup B Neisseria meningitidis, which has been developed using reverse vaccinology and is the basis of a meningococcal B vaccine entering phase III clinical trials. Binding of factor H (fH), an inhibitor of the complement alternative pathway, to fHBP enables N. meningitidis to evade killing by the innate immune system. All fHBP null mutant strains analyzed were sensitive to killing in ex vivo human whole blood and serum models of meningococcal bacteremia with respect to the isogenic wild-type strains. The fHBP mutant strains of MC58 and BZ83 (high fHBP expressors) survived in human blood and serum for less than 60 min (decrease of >2 log10 CFU), while NZ98/254 (intermediate fHBP expressor) and 67/00 (low fHBP expressor) showed decreases of >1 log10 CFU after 60 to 120 min of incubation. In addition, fHBP is important for survival in the presence of the antimicrobial peptide LL-37 (decrease of >3 log10 CFU after 2 h of incubation), most likely due to electrostatic interactions between fHBP and the cationic LL-37 molecule. Hence, the expression of fHBP by N. meningitidis strains is important for survival in human blood and human serum and in the presence of LL-37, even at low levels. The functional significance of fHBP in mediating resistance to the human immune response, in addition to its widespread distribution and its ability to induce bactericidal antibodies, indicates that it is an important component of the serogroup B meningococcal vaccine.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Defining a protective epitope on factor H binding protein, a key meningococcal virulence factor and vaccine antigen

Enrico Malito; Agnese Faleri; Paola Lo Surdo; Daniele Veggi; Giulietta Maruggi; Eva Grassi; Elena Cartocci; Isabella Bertoldi; Alessia Genovese; Laura Santini; Giacomo Romagnoli; Erica Borgogni; Sébastien Brier; Carla Lo Passo; Maria Domina; Flora Castellino; Franco Felici; Stijn van der Veen; Steven Johnson; Susan M. Lea; Christoph M. Tang; Mariagrazia Pizza; Silvana Savino; Nathalie Norais; Rino Rappuoli; Matthew J. Bottomley; Vega Masignani

Mapping of epitopes recognized by functional monoclonal antibodies (mAbs) is essential for understanding the nature of immune responses and designing improved vaccines, therapeutics, and diagnostics. In recent years, identification of B-cell epitopes targeted by neutralizing antibodies has facilitated the design of peptide-based vaccines against highly variable pathogens like HIV, respiratory syncytial virus, and Helicobacter pylori; however, none of these products has yet progressed into clinical stages. Linear epitopes identified by conventional mapping techniques only partially reflect the immunogenic properties of the epitope in its natural conformation, thus limiting the success of this approach. To investigate antigen–antibody interactions and assess the potential of the most common epitope mapping techniques, we generated a series of mAbs against factor H binding protein (fHbp), a key virulence factor and vaccine antigen of Neisseria meningitidis. The interaction of fHbp with the bactericidal mAb 12C1 was studied by various epitope mapping methods. Although a 12-residue epitope in the C terminus of fHbp was identified by both Peptide Scanning and Phage Display Library screening, other approaches, such as hydrogen/deuterium exchange mass spectrometry (MS) and X-ray crystallography, showed that mAb 12C1 occupies an area of ∼1,000 Å2 on fHbp, including >20 fHbp residues distributed on both N- and C-terminal domains. Collectively, these data show that linear epitope mapping techniques provide useful but incomplete descriptions of B-cell epitopes, indicating that increased efforts to fully characterize antigen–antibody interfaces are required to understand and design effective immunogens.


Molecular Microbiology | 2006

Neisseria meningitidis NhhA is a multifunctional trimeric autotransporter adhesin

Maria Scarselli; Davide Serruto; Paolo Montanari; Barbara Capecchi; Jeannette Adu-Bobie; Daniele Veggi; Rino Rappuoli; Mariagrazia Pizza; Beatrice Aricò

NhhA, Neisseriahia/hsf homologue, or GNA0992, is an oligomeric outer membrane protein of Neisseria meningitidis, recently included in the family of trimeric autotransporter adhesins. In this study we present the structural and functional characterization of this protein. By expressing in Escherichia coli the full‐length gene, deletion mutants and chimeric proteins of NhhA, we demonstrated that the last 72 C‐terminal residues are able to allow trimerization and localization of the N‐terminal protein domain to the bacterial surface. In addition, we investigated on the possible role of NhhA in bacterial–host interaction events. We assessed in vitro the ability of recombinant purified NhhA to bind human epithelial cells as well as laminin and heparan sulphate. Furthermore, we shown that E. coli strain expressing NhhA was able to adhere to epithelial cells, and observed a reduced adherence in a meningococcal isogenic MC58ΔNhhA mutant. We concluded that this protein is a multifunctional adhesin, able to promote the bacterial adhesion to host cells and extracellular matrix components. Collectively, our results underline a putative role of NhhA in meningococcal pathogenesis and ascertain its structural and functional belonging to the emerging group of bacterial autotransporter adhesins with trimeric architecture.


Molecular Microbiology | 2003

Neisseria meningitidis App, a new adhesin with autocatalytic serine protease activity

Davide Serruto; Jeannette Adu-Bobie; Maria Scarselli; Daniele Veggi; Mariagrazia Pizza; Rino Rappuoli; Beatrice Aricò

Neisseria meningitidis is a Gram‐negative bacterium which colonizes the human upper respiratory tract. Occasionally, it translocates to the bloodstream causing sepsis and from there it can cross the blood–brain barrier and cause meningitis. Many of the molecules, which mediate the interaction of N. meningitidis to host cells, are still unknown. Recently, App ( A dhesion and p enetration p rotein) was described as a member of the autotransporter family and a homologue to the Hap ( H aemophilus a dhesion and p enetration) protein of Haemophilus influenzae , a molecule that plays a role in the interaction with human epithelial cells. In this study we expressed app in Escherichia coli in order to analyse the functional properties of the protein. We show that the protein is exported to the E. coli surface, processed by an endogenous serine‐protease activity and released in the culture supernatant. Escherichia coli expressing app adhere to Chang epithelial cells, showing that App is able to mediate bacterial adhesion to host cells. The serine protease activity is localized at the amino‐terminal domain, whereas the binding domain is in the carboxy‐terminal region. The role of App in adhesion was confirmed also in N. meningitidis .


Journal of Biological Chemistry | 2006

Solution structure of the immunodominant domain of protective antigen GNA1870 of Neisseria meningitidis.

Francesca Cantini; Silvana Savino; Maria Scarselli; Vega Masignani; Mariagrazia Pizza; Giacomo Romagnoli; Erwin Swennen; Daniele Veggi; Lucia Banci; Rino Rappuoli

GNA1870, a 28-kDa surface-exposed lipoprotein of Neisseria meningitidis recently discovered by reverse vaccinology, is one of the most potent antigens of Meningococcus and a promising candidate for a universal vaccine against a devastating disease. Previous studies of epitope mapping and genetic characterization identified residues critical for bactericidal response within the C-terminal domain of the molecule. To elucidate the conformation of protective epitopes, we used NMR spectroscopy to obtain the solution structure of the immunodominant 18-kDa C-terminal portion of GNA1870. The structure consists of an eight-stranded antiparallel β-barrel overlaid by a short α-helix with an unstructured N-terminal end. Residues previously shown to be important for antibody recognition were mapped on loops facing the same ridge of the molecule. The sequence similarity of GNA1870 with members of the bacterial transferrin receptor family allows one to predict the folding of this class of well known bacterial antigens, providing the basis for the rational engineering of high affinity B cell epitopes.


Journal of Biological Chemistry | 2009

Solution structure of the factor H binding protein, a survival factor and protective antigen of Neisseria meningitidis

Francesca Cantini; Daniele Veggi; Sara Dragonetti; Silvana Savino; Maria Scarselli; Giacomo Romagnoli; Mariagrazia Pizza; Lucia Banci; Rino Rappuoli

Factor H-binding protein is a 27-kDa lipoprotein of Neisseria meningitidis discovered while screening the bacterial genome for vaccine candidates. In addition to being an important component of a vaccine against meningococcus in late stage of development, the protein is essential for pathogenesis because it allows the bacterium to survive and grow in human blood by binding the human complement factor H. We recently reported the solution structure of the C-terminal domain of factor H-binding protein, which contains the immunodominant epitopes. In the present study, we report the structure of the full-length molecule, determined by nuclear magnetic resonance spectroscopy. The protein is composed of two independent barrels connected by a short link. Mapping the residues recognized by monoclonal antibodies with bactericidal or factor H binding inhibition properties allowed us to predict the sites involved in the function of the protein. The structure therefore provides the basis for designing improved vaccine molecules.


Journal of Molecular Biology | 2009

Epitope Mapping of a Bactericidal Monoclonal Antibody against the Factor H Binding Protein of Neisseria meningitidis

Maria Scarselli; Francesca Cantini; Laura Santini; Daniele Veggi; Sara Dragonetti; Claudio Donati; Silvana Savino; Marzia Monica Giuliani; Maurizio Comanducci; Federica Di Marcello; Giacomo Romagnoli; Mariagrazia Pizza; Lucia Banci; Rino Rappuoli

The factor H binding protein (fHbp) is a 27-kDa membrane-anchored lipoprotein of Neisseria meningitidis that allows the survival of the bacterium in human plasma; it is also a major component of a universal vaccine against meningococcus B. In this study, we used nuclear magnetic resonance spectroscopy, mutagenesis, and in silico modeling to map the epitope recognized by MAb502, a bactericidal monoclonal antibody elicited by fHbp. The data show that the antibody recognizes a conformational epitope within a well-defined area of the immunodominant C-terminal domain of the protein that is formed by two loops connecting different beta-strands of a beta-barrel and a short alpha-helix brought in spatial proximity by the protein folding. The identification of the protective epitopes of fHbp is an important factor for understanding the mechanism(s) of an effective immune response and provides valuable guidelines for designing variants of the protein able to induce broadly protective immunity.


Mbio | 2012

FdeC, a Novel Broadly Conserved Escherichia coli Adhesin Eliciting Protection against Urinary Tract Infections

Barbara Nesta; Glen Spraggon; Christopher J. Alteri; Danilo Gomes Moriel; Roberto Rosini; Daniele Veggi; Sara N. Smith; Isabella Bertoldi; Ilaria Pastorello; Ilaria Ferlenghi; Maria Rita Fontana; Gad Frankel; Harry L. T. Mobley; Rino Rappuoli; Mariagrazia Pizza; Laura Serino; Marco Soriani

ABSTRACT The increasing antibiotic resistance of pathogenic Escherichia coli species and the absence of a pan-protective vaccine pose major health concerns. We recently identified, by subtractive reverse vaccinology, nine Escherichia coli antigens that protect mice from sepsis. In this study, we characterized one of them, ECOK1_0290, named FdeC (factor adherence E. coli) for its ability to mediate E. coli adhesion to mammalian cells and extracellular matrix. This adhesive propensity was consistent with the X-ray structure of one of the FdeC domains that shows a striking structural homology to Yersinia pseudotuberculosis invasin and enteropathogenic E. coli intimin. Confocal imaging analysis revealed that expression of FdeC on the bacterial surface is triggered by interaction of E. coli with host cells. This phenotype was also observed in bladder tissue sections derived from mice infected with an extraintestinal strain. Indeed, we observed that FdeC contributes to colonization of the bladder and kidney, with the wild-type strain outcompeting the fdeC mutant in cochallenge experiments. Finally, intranasal mucosal immunization with recombinant FdeC significantly reduced kidney colonization in mice challenged transurethrally with uropathogenic E. coli, supporting a role for FdeC in urinary tract infections. IMPORTANCE Pathogenic Escherichia coli strains are involved in a diverse spectrum of diseases, including intestinal and extraintestinal infections (urinary tract infections and sepsis). The absence of a broadly protective vaccine against all these E. coli strains is a major problem for modern society due to high costs to health care systems. Here, we describe the structural and functional properties of a recently reported protective antigen, named FdeC, and elucidated its putative role during extraintestinal pathogenic E. coli infection by using both in vitro and in vivo infection models. The conservation of FdeC among strains of different E. coli pathotypes highlights its potential as a component of a broadly protective vaccine against extraintestinal and intestinal E. coli infections. Pathogenic Escherichia coli strains are involved in a diverse spectrum of diseases, including intestinal and extraintestinal infections (urinary tract infections and sepsis). The absence of a broadly protective vaccine against all these E. coli strains is a major problem for modern society due to high costs to health care systems. Here, we describe the structural and functional properties of a recently reported protective antigen, named FdeC, and elucidated its putative role during extraintestinal pathogenic E. coli infection by using both in vitro and in vivo infection models. The conservation of FdeC among strains of different E. coli pathotypes highlights its potential as a component of a broadly protective vaccine against extraintestinal and intestinal E. coli infections.


Molecular Microbiology | 2003

NarE: a novel ADP-ribosyltransferase from Neisseria meningitidis.

Vega Masignani; Enrico Balducci; Federica Di Marcello; Silvana Savino; Davide Serruto; Daniele Veggi; Stefania Bambini; Maria Scarselli; Beatrice Aricò; Maurizio Comanducci; Jeannette Adu-Bobie; Marzia Monica Giuliani; Rino Rappuoli; Mariagrazia Pizza

Mono ADP‐ribosyltransferases (ADPRTs) are a class of functionally conserved enzymes present in prokaryotic and eukaryotic organisms. In bacteria, these enzymes often act as potent toxins and play an important role in pathogenesis. Here we report a profile‐based computational approach that, assisted by secondary structure predictions, has allowed the identification of a previously undiscovered ADP‐ribosyltransferase in Neisseria meningitidis (NarE). NarE shows structural homologies with E. coli heat‐labile enterotoxin (LT) and cholera toxin (CT) and possesses ADP‐ribosylating and NAD‐glycohydrolase activities. As in the case of LT and CT, NarE catalyses the transfer of the ADP‐ribose moiety to arginine residues. Despite the absence of a signal peptide, the protein is efficiently exported into the periplasm of Neisseria. The narE gene is present in 25 out of 43 strains analysed, is always present in ET‐5 and Lineage 3 but absent in ET‐37 and Cluster A4 hypervirulent lineages. When present, the gene is 100% conserved in sequence and is inserted upstream of and co‐transcribed with the lipoamide dehydrogenase E3 gene. Possible roles in the pathogenesis of N. meningitidis are discussed.

Collaboration


Dive into the Daniele Veggi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lucia Banci

University of Florence

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge