Network


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

Hotspot


Dive into the research topics where Francesca Buricchi is active.

Publication


Featured researches published by Francesca Buricchi.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Approach to discover T- and B-cell antigens of intracellular pathogens applied to the design of Chlamydia trachomatis vaccines

Oretta Finco; Elisabetta Frigimelica; Francesca Buricchi; Roberto Petracca; Giuliano Galli; Elisa Faenzi; Eva Meoni; Alessandra Bonci; Mauro Agnusdei; Filomena Nardelli; Erika Bartolini; Maria Scarselli; Elena Caproni; Donatello Laera; Luisanna Zedda; David Skibinski; Serena Giovinazzi; Riccardo Bastone; Elvira Ianni; Roberto Cevenini; Guido Grandi; Renata Grifantini

Natural immunity against obligate and/or facultative intracellular pathogens is usually mediated by both humoral and cellular immunity. The identification of those antigens stimulating both arms of the immune system is instrumental for vaccine discovery. Although high-throughput technologies have been applied for the discovery of antibody-inducing antigens, few examples of their application for T-cell antigens have been reported. We describe how the compilation of the immunome, here defined as the pool of immunogenic antigens inducing T- and B-cell responses in vivo, can lead to vaccine candidates against Chlamydia trachomatis. We selected 120 C. trachomatis proteins and assessed their immunogenicity using two parallel high-throughput approaches. Protein arrays were generated and screened with sera from C. trachomatis-infected patients to identify antibody-inducing antigens. Splenocytes from C. trachomatis-infected mice were stimulated with 79 proteins, and the frequency of antigen-specific CD4+/IFN-γ+ T cells was analyzed by flow cytometry. We identified 21 antibody-inducing antigens, 16 CD4+/IFN-γ+–inducing antigens, and five antigens eliciting both types of responses. Assessment of their protective activity in a mouse model of Chlamydia muridarum lung infection led to the identification of seven antigens conferring partial protection when administered with LTK63/CpG adjuvant. Protection was largely the result of cellular immunity as assessed by CD4+ T-cell depletion. The seven antigens provided robust additive protection when combined in four-antigen combinations. This study paves the way for the development of an effective anti-Chlamydia vaccine and provides a general approach for the discovery of vaccines against other intracellular pathogens.


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

Human circulating influenza-CD4+ ICOS1+IL-21+ T cells expand after vaccination, exert helper function, and predict antibody responses

Fabiana Spensieri; Erica Borgogni; Luisanna Zedda; Monia Bardelli; Francesca Buricchi; Gianfranco Volpini; Elena Fragapane; Simona Tavarini; Oretta Finco; Rino Rappuoli; Giuseppe Del Giudice; Grazia Galli; Flora Castellino

Protection against influenza is mediated by neutralizing antibodies, and their induction at high and sustained titers is key for successful vaccination. Optimal B cells activation requires delivery of help from CD4+ T lymphocytes. In lymph nodes and tonsils, T-follicular helper cells have been identified as the T cells subset specialized in helping B lymphocytes, with interleukin-21 (IL-21) and inducible costimulatory molecule (ICOS1) playing a central role for this function. We followed the expansion of antigen-specific IL-21+ CD4+ T cells upon influenza vaccination in adults. We show that, after an overnight in vitro stimulation, influenza-specific IL-21+ CD4+ T cells can be measured in human blood, accumulate in the CXCR5−ICOS1+ population, and increase in frequency after vaccination. The expansion of influenza-specific ICOS1+IL-21+ CD4+ T cells associates with and predicts the rise of functionally active antibodies to avian H5N1. We also show that blood-derived CXCR5−ICOS1+ CD4+ T cells exert helper function in vitro and support the differentiation of influenza specific B cells in an ICOS1- and IL-21–dependent manner. We propose that the expansion of antigen-specific ICOS1+IL-21+ CD4+ T cells in blood is an early marker of vaccine immunogenicity and an important immune parameter for the evaluation of novel vaccination strategies.


Infection and Immunity | 2009

CT043, a protective antigen that induces a CD4+ Th1 response during Chlamydia trachomatis infection in mice and humans.

Eva Meoni; Elisa Faenzi; Elisabetta Frigimelica; Luisanna Zedda; David Skibinski; Serena Giovinazzi; Alessandra Bonci; Roberto Petracca; Erika Bartolini; Giuliano Galli; Mauro Agnusdei; Filomena Nardelli; Francesca Buricchi; Nathalie Norais; Ilaria Ferlenghi; Manuela Donati; Roberto Cevenini; Oretta Finco; Guido Grandi; Renata Grifantini

ABSTRACT Despite several decades of intensive studies, no vaccines against Chlamydia trachomatis, an intracellular pathogen causing serious ocular and urogenital diseases, are available yet. Infection-induced immunity in both animal models and humans strongly supports the notion that for a vaccine to be effective a strong CD4+ Th1 immune response should be induced. In the course of our vaccine screening program based on the selection of chlamydial proteins eliciting cell-mediated immunity, we have found that CT043, a protein annotated as hypothetical, induces CD4+ Th1 cells both in chlamydia-infected mice and in human patients with diagnosed C. trachomatis genital infection. DNA priming/protein boost immunization with CT043 results in a 2.6-log inclusion-forming unit reduction in the murine lung infection model. Sequence analysis of CT043 from C. trachomatis human isolates belonging to the most representative genital serovars revealed a high degree of conservation, suggesting that this antigen could provide cross-serotype protection. Therefore, CT043 is a promising vaccine candidate against C. trachomatis infection.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Expression of factor H binding protein in meningococcal strains can vary at least 15-fold and is genetically determined

Massimiliano Biagini; Marco Spinsanti; Gabriella De Angelis; Ilaria Ferlenghi; Maria Scarselli; Fabio Rigat; Nicola Messuti; Alessia Biolchi; Alessandro Muzzi; Giulia Anderloni; Brunella Brunelli; Elena Cartocci; Francesca Buricchi; Chiara Tani; Maria Stella; Monica Moschioni; Elena Del Tordello; Annalisa Colaprico; Silvana Savino; Marzia Monica Giuliani; Isabel Delany; Mariagrazia Pizza; Paolo Costantino; Nathalie Norais; Rino Rappuoli; Vega Masignani

Significance Complement is the main line of defense against bacterial pathogens; however, the molecular mechanisms triggering killing are largely unknown. Factor H binding protein (fHbp) is a component of two licensed vaccines against serogroup B meningococcus and a key target of complement-mediated bacterial killing. Selected reaction monitoring was used for the absolute quantification of fHbp on invasive meningococcal strains, showing that expression among strains can vary at least 15-fold and a minimum of 757 molecules separated by not more than 130 nm are required to engage C1q and kill the bacteria. Furthermore, the amount of fHbp is genetically determined by the sequence of the promoter region and correlated with the bactericidal activity. These findings increase the understanding of complement-mediated killing and vaccine protection. Factor H binding protein (fHbp) is a lipoprotein of Neisseria meningitidis important for the survival of the bacterium in human blood and a component of two recently licensed vaccines against serogroup B meningococcus (MenB). Based on 866 different amino acid sequences this protein is divided into three variants or two families. Quantification of the protein is done by immunoassays such as ELISA or FACS that are susceptible to the sequence variation and expression level of the protein. Here, selected reaction monitoring mass spectrometry was used for the absolute quantification of fHbp in a large panel of strains representative of the population diversity of MenB. The analysis revealed that the level of fHbp expression can vary at least 15-fold and that variant 1 strains express significantly more protein than variant 2 or variant 3 strains. The susceptibility to complement-mediated killing correlated with the amount of protein expressed by the different meningococcal strains and this could be predicted from the nucleotide sequence of the promoter region. Finally, the absolute quantification allowed the calculation of the number of fHbp molecules per cell and to propose a mechanistic model of the engagement of C1q, the recognition component of the complement cascade.


Vaccine | 2012

One dose of an MF59-adjuvanted pandemic A/H1N1 vaccine recruits pre-existing immune memory and induces the rapid rise of neutralizing antibodies.

Elisa Faenzi; Luisanna Zedda; Monia Bardelli; Fabiana Spensieri; Erica Borgogni; Gianfranco Volpini; Francesca Buricchi; Franco Laghi Pasini; Pier Leopoldo Capecchi; Fabio Montanaro; Riccardo Belli; Maria Lattanzi; Simona Piccirella; Emanuele Montomoli; Syed Sohail Ahmed; Rino Rappuoli; Giuseppe Del Giudice; Oretta Finco; Flora Castellino; Grazia Galli

Protective antibody responses to a single dose of 2009 pandemic vaccines have been observed in the majority of healthy subjects aged more than 3 years. These findings suggest that immune memory lymphocytes primed by previous exposure to seasonal influenza antigens are recruited in the response to A/H1N1 pandemic vaccines and allow rapid seroconversion. However, a clear dissection of the immune memory components favoring a fast response to pandemic vaccination is still lacking. Here we report the results from a clinical study where antibody, CD4+ T cell, plasmablast and memory B cell responses to one dose of an MF59-adjuvanted A/H1N1 pandemic vaccine were analyzed in healthy adults. While confirming the rapid appearance of antibodies neutralizing the A/H1N1 pandemic virus, we show here that the response is dominated by IgG-switched antibodies already in the first week after vaccination. In addition, we found that vaccination induces the rapid expansion of pre-existing CD4+ T cells and IgG-memory B lymphocytes cross-reactive to seasonal and pandemic A/H1N1 antigens. These data shed light on the different components of the immune response to the 2009 H1N1 pandemic influenza vaccination and may have implications in the design of vaccination strategies against future influenza pandemics.


The FASEB Journal | 2014

Two cross-reactive monoclonal antibodies recognize overlapping epitopes on Neisseria meningitidis factor H binding protein but have different functional properties

Agnese Faleri; Laura Santini; Sébastien Brier; Werner Pansegrau; Paola Lo Surdo; Maria Scarselli; Francesca Buricchi; Gianfranco Volpini; Alessia Genovese; Stijn van der Veen; Susan M. Lea; Christoph M. Tang; Silvana Savino; Mariagrazia Pizza; Oretta Finco; Nathalie Norais; Vega Masignani

Factor H binding protein (fHbp) is one of the main antigens of the 4‐component meningococcus B (4CMenB) multicomponent vaccine against disease caused by serogroup B Neisseria meningitidis (MenB). fHbp binds the complement down‐regulating protein human factor H (hfH), thus resulting in immune evasion. fHbp exists in 3 variant groups with limited cross‐protective responses. Previous studies have described the generation of monoclonal antibodies (mAbs) targeting variant‐specific regions of fHbp. Here we report for the first time the functional characterization of two mAbs that recognize a wide panel of fHbp variants and subvariants on the MenB surface and that are able to inhibit fHbp binding to hfH. The antigenic regions targeted by the two mAbs were accurately mapped by hydrogen‐deuterium exchange mass spectrometry (HDX‐MS), revealing partially overlapping epitopes on the N terminus of fHbp. Furthermore, while none of the mAbs had bactericidal activity on its own, a synergistic effect was observed for each of them when tested by the human complement serum bactericidal activity (hSBA) assay in combination with a second nonbactericidal mAb. The bases underlying fHbp variant cross‐reactivity, as well as inhibition of hfH binding and cooperativity effect observed for the two mAbs, are discussed in light of the mapped epitopes.—Faleri, A, Santini, L., Brier, S., Pansegrau, W., Lo Surdo, P., Scarselli, M., Buricchi, F., Volpini, G., Genovese, A, van der Veen, S., Lea, S., Tang, C. M., Savino, S., Pizza, M., Finco, O., Norais, N., Masignani, V Two cross‐reactive monoclonal antibodies recognize overlapping epitopes on Neisseria meningitidis factor H binding protein but have different functional properties. FASEB J. 28, 28–1644 (1653). www.fasebj.org


European Journal of Immunology | 2013

Impact of preexisting memory to seasonal A/H1N1 influenza virus on the immune response following vaccination against avian A/H5N1 virus

Francesca Buricchi; Monia Bardelli; Carmine Malzone; Barbara Capecchi; Uwe Nicolay; Elena Fragapane; Flora Castellino; Giuseppe Del Giudice; Grazia Galli; Oretta Finco

Cross‐protection against divergent strains of influenza virus is an objective of various vaccination approaches. B cells cross‐neutralizing several influenza A heterosubtypes have been isolated from cultured human memory B cells (MBCs) and plasmablasts early after influenza vaccination or infection. However, a systematic assessment of the frequency of MBCs and plasmablasts in the blood of healthy individuals is lacking. Here, we show that under resting conditions about 45% of human adults never vaccinated nor exposed to avian A/H5N1 influenza have detectable circulating MBCs cross‐reacting with H5N1. This proportion rises to 63.3% among subjects with a large pool of MBCs specific for seasonal H1N1 (i.e. frequency ≥1% of total IgG MBCs). Moreover, subjects with high baseline frequencies of H1N1‐specific MBCs had an expansion of H5N1‐specific MBCs producing H5‐neutralizing antibodies already after the first dose of an MF59‐adjuvanted H5N1 vaccine. These results suggest that H1N1‐specific MBCs contain a subset of cells cross‐reacting to H5. We propose that a proportion of human adults have a pool of H5/H1 cross‐reactive MBCs that contribute to the rapid rise of the antibody response to divergent influenza strains. This may have implications on vaccination strategies aimed at counteracting future influenza pandemics.


PLOS ONE | 2013

Ex Vivo Analysis of Human Memory B Lymphocytes Specific for A and B Influenza Hemagglutinin by Polychromatic Flow-Cytometry

Monia Bardelli; Liliana Alleri; Francesca Angiolini; Francesca Buricchi; Simona Tavarini; Chiara Sammicheli; Sandra Nuti; Elena Degl'Innocenti; Isabelle Isnardi; Elena Fragapane; Giuseppe Del Giudice; Flora Castellino; Grazia Galli

Understanding the impact that human memory B-cells (MBC), primed by previous infections or vaccination, exert on neutralizing antibody responses against drifted influenza hemagglutinin (HA) is key to design best protective vaccines. A major obstacle to these studies is the lack of practical tools to analyze HA-specific MBCs in human PBMCs ex vivo. We report here an efficient method to identify MBCs carrying HA-specific BCR in frozen PBMC samples. By using fluorochrome-tagged recombinant HA baits, and vaccine antigens from mismatched influenza strains to block BCR-independent binding, we developed a protocol suitable for quantitative, functional and molecular analysis of single MBCs specific for HA from up to two different influenza strains in the same tube. This approach will permit to identify the naive and MBC precursors of plasmablasts and novel MBCs appearing in the blood following infection or vaccination, thus clarifying the actual contribution of pre-existing MBCs in antibody responses against novel influenza viruses. Finally, this protocol can allow applying high throughput deep sequencing to analyze changes in the repertoire of HA+ B-cells in longitudinal samples from large cohorts of vaccinees and infected subjects with the ultimate goal of understanding the in vivo B-cell dynamics driving the evolution of broadly cross-protective antibody responses.


Scientific Reports | 2018

Human protective response induced by meningococcus B vaccine is mediated by the synergy of multiple bactericidal epitopes

M. Giuliani; Erika Bartolini; Barbara Galli; Laura Santini; P. Lo Surdo; Francesca Buricchi; M. Bruttini; B. Benucci; N. Pacchiani; L. Alleri; D. Donnarumma; Werner Pansegrau; I. Peschiera; Ilaria Ferlenghi; Roberta Cozzi; Nathalie Norais; Marzia Monica Giuliani; Domenico Maione; M Pizza; Rino Rappuoli; Oretta Finco; Vega Masignani

Abstract4CMenB is the first broad coverage vaccine for the prevention of invasive meningococcal disease caused by serogroup B strains. To gain a comprehensive picture of the antibody response induced upon 4CMenB vaccination and to obtain relevant translational information directly from human studies, we have isolated a panel of human monoclonal antibodies from adult vaccinees. Based on the Ig-gene sequence of the variable region, 37 antigen-specific monoclonal antibodies were identified and produced as recombinant Fab fragments, and a subset also produced as full length recombinant IgG1 and functionally characterized. We found that the monoclonal antibodies were cross-reactive against different antigen variants and recognized multiple epitopes on each of the antigens. Interestingly, synergy between antibodies targeting different epitopes enhanced the potency of the bactericidal response. This work represents the first extensive characterization of monoclonal antibodies generated in humans upon 4CMenB immunization and contributes to further unraveling the immunological and functional properties of the vaccine antigens. Moreover, understanding the mechanistic nature of protection induced by vaccination paves the way to more rational vaccine design and implementation.


Immunity, inflammation and disease | 2013

Optimized fluorescent labeling to identify memory B cells specific for Neisseria meningitidis serogroup B vaccine antigens ex vivo

Nitya Nair; Ludovico Buti; Elisa Faenzi; Francesca Buricchi; Sandra Nuti; Chiara Sammicheli; Simona Tavarini; Maximilian Wl Popp; Hidde L. Ploegh; Francesco Berti; Mariagrazia Pizza; Flora Castellino; Oretta Finco; Rino Rappuoli; Giuseppe Del Giudice; Grazia Galli; Monia Bardelli

Antigen‐specific memory B cells generate anamnestic responses and high affinity antibodies upon re‐exposure to pathogens. Attempts to isolate rare antigen‐specific memory B cells for in‐depth functional analysis at the single‐cell level have been hindered by the lack of tools with adequate sensitivity. We applied two independent methods of protein labeling to sensitive and specific ex vivo identification of antigen‐specific memory B cells by flow cytometry: stringently controlled amine labeling, and sortagging, a novel method whereby a single nucleophilic fluorochrome molecule is added onto an LPETG motif carried by the target protein. We show that sortagged NadA, a major antigen in the meningococcal serogroup B vaccine, identifies NadA‐specific memory B cells with high sensitivity and specificity, comparable to NadA amine‐labeled under stringent reaction parameters in a mouse model of vaccination. We distinguish NadA‐specific switched MBC induced by vaccination from the background signal contributed by splenic transitional and marginal zone B cells. In conclusion, we demonstrate that protein structural data coupled with sortag technology allows the development of engineered antigens that are as sensitive and specific as conventional chemically labeled antigens in detecting rare MBC, and minimize the possibility of disrupting conformational B cell epitopes.

Collaboration


Dive into the Francesca Buricchi's collaboration.

Researchain Logo
Decentralizing Knowledge