Network


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

Hotspot


Dive into the research topics where Francesca Schena is active.

Publication


Featured researches published by Francesca Schena.


Stem Cells | 2008

Bone Marrow‐Derived Mesenchymal Stem Cells Induce Both Polyclonal Expansion and Differentiation of B Cells Isolated from Healthy Donors and Systemic Lupus Erythematosus Patients

Elisabetta Traggiai; Stefano Volpi; Francesca Schena; Marco Gattorno; Francesca Ferlito; Lorenzo Moretta; Alberto Martini

Human bone marrow multipotent mesenchymal stromal cells are progenitor cells that can be expanded in vitro and differentiate into various cells of mesodermal origin. They contribute to the bone marrow reticular niche, where mature B cells and long‐lived plasma cells are maintained. Multipotent mesenchymal stromal cells were recently shown to modulate T‐ and B‐cell proliferation and differentiation, dendritic cell maturation, and natural killer activity. These immunoregulatory properties encouraged a possible use of these cells to modulate autoimmune responses in humans. We studied the influence of bone marrow mesenchymal stem cells on highly purified B‐cell subsets isolated from healthy donors and total B cells from pediatric systemic lupus erythematosus patients. Bone marrow mesenchymal stem cells promoted proliferation and differentiation into immunoglobulin‐secreting cells of transitional and naïve B cells stimulated with an agonist of Toll‐like receptor 9, in the absence of B cell receptor triggering. They strongly enhanced proliferation and differentiation into plasma cells of memory B‐cell populations. A similar effect was observed in response to polyclonal stimulation of B cells isolated from pediatric patients with systemic lupus erythematosus. This study casts important questions on bone marrow mesenchymal stem cells as a therapeutic tool in autoimmune diseases in which B‐cell activation is crucially implicated in the pathogenesis of the disease.


Arthritis & Rheumatism | 2010

Interferon‐γ–dependent inhibition of B cell activation by bone marrow–derived mesenchymal stem cells in a murine model of systemic lupus erythematosus

Francesca Schena; Claudio Gambini; Andrea Gregorio; Manuela Mosconi; Daniele Reverberi; Marco Gattorno; Simona Casazza; Antonio Uccelli; Lorenzo Moretta; Alberto Martini; Elisabetta Traggiai

OBJECTIVE Bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent cells characterized by immunomodulatory properties and are therefore considered a promising tool for the treatment of immune-mediated diseases. This study was undertaken to assess the influence of murine BM-MSCs on the activation of B cells in (NZB × NZW)F(1) mice as an animal model of systemic lupus erythematosus (SLE). METHODS We evaluated the in vitro effects of BM-MSCs on the proliferation and differentiation to plasma cells of splenic mature B cell subsets, namely follicular and marginal zone B cells isolated from (NZB × NZW)F(1) mice. Lupus mice were also treated with BM-MSCs, and serum autoantibodies, proteinuria, histologic changes in the kidney, and survival rates were monitored. RESULTS BM-MSCs inhibited antigen-dependent proliferation and differentiation to plasma cells of follicular and marginal zone B cells in vitro. This inhibitory effect was dependent on interferon-γ (IFNγ) and was mediated by cell-to-cell contact, involving the programmed death 1 (PD-1)/PD ligand pathway. In vivo treatment with BM-MSCs did not affect the levels of anti-double-stranded DNA antibodies or proteinuria. However, a reduction in glomerular immune complex deposition, lymphocytic infiltration, and glomerular proliferation was observed. CONCLUSION Our findings indicate that BM-MSCs affect B cell receptor-dependent activation of both follicular and marginal zone B cells from lupus mice. This inhibitory effect is IFNγ-dependent and cell contact-dependent. MSCs in vivo do not affect the production of autoantibodies, the level of proteinuria, or the mortality rates. Nonetheless, the significant improvement in histologic findings in the kidney supports the potential role of MSCs in the prevention of glomerular damage.


Journal of Experimental Medicine | 2010

Homeostatic expansion of autoreactive immunoglobulin-secreting cells in the Rag2 mouse model of Omenn syndrome

Barbara Cassani; Pietro Luigi Poliani; Veronica Marrella; Francesca Schena; Aisha V. Sauer; Maria Ravanini; Dario Strina; Christian E. Busse; Stephan Regenass; Hedda Wardemann; Alberto Martini; Fabio Facchetti; Mirjam van der Burg; Antonius Rolink; Paolo Vezzoni; Fabio Grassi; Elisabetta Traggiai; Anna Villa

Hypomorphic RAG mutations, leading to limited V(D)J rearrangements, cause Omenn syndrome (OS), a peculiar severe combined immunodeficiency associated with autoimmune-like manifestations. Whether B cells play a role in OS pathogenesis is so far unexplored. Here we report the detection of plasma cells in lymphoid organs of OS patients, in which circulating B cells are undetectable. Hypomorphic Rag2R229Q knock-in mice, which recapitulate OS, revealed, beyond severe B cell developmental arrest, a normal or even enlarged compartment of immunoglobulin-secreting cells (ISC). The size of this ISC compartment correlated with increased expression of Blimp1 and Xbp1, and these ISC were sustained by elevated levels of T cell derived homeostatic and effector cytokines. The detection of high affinity pathogenic autoantibodies toward target organs indicated defaults in B cell selection and tolerance induction. We hypothesize that impaired B cell receptor (BCR) editing and a serum B cell activating factor (BAFF) abundance might contribute toward the development of a pathogenic B cell repertoire in hypomorphic Rag2R229Q knock-in mice. BAFF-R blockade reduced serum levels of nucleic acid-specific autoantibodies and significantly ameliorated inflammatory tissue damage. These findings highlight a role for B cells in OS pathogenesis.


Human Heredity | 2007

Genetic predisposition to familial neuroblastoma: Identification of two novel genomic regions at 2p and 12p

Luca Longo; Emanuele Panza; Francesca Schena; Marco Seri; Marcella Devoto; Giovanni Romeo; Carla Bini; G. Pappalardo; Gian Paolo Tonini; Patrizia Perri

Objectives: The rarity of familial neuroblastoma (NB) has allowed only a few linkage studies, most of which did not show any evidence of linkage to regions involved in somatic alterations or to genes implicated in other neurocristopathies seldom associated with NB. We screened a highly informative family with recurrent NB by genome-wide linkage analysis aimed at identifying chromosomal regions for NB predisposing genes. Methods: A genome-wide screen was performed using 382 microsatellite markers. Multipoint model-based linkage analysis was carried out under a dominant mode of inheritance for the disease using the ‘affected only’ approach. Results: Our analysis identified two haplotypes co-segregating with the disease on chromosomes 2p and 12p, and yielded maximum lod-score values of 3.01 (p < 0.0001) for markers on both intervals. Conclusions: Evidence of linkage was reported at 16p in North American families, whereas our studies excluded this interval and indicated other loci for disease predisposition, thus confirming the remarkable genetic heterogeneity of NB. These results suggest an oligogenic inheritance in NB involving more loci in genetic determination of the disease.


The Journal of Allergy and Clinical Immunology | 2011

Lentiviral-mediated gene therapy leads to improvement of B-cell functionality in a murine model of Wiskott-Aldrich syndrome

Marita Bosticardo; Elena Draghici; Francesca Schena; Aisha V. Sauer; Elena Fontana; Maria Carmina Castiello; Marco Catucci; Michela Locci; Luigi Naldini; Alessandro Aiuti; Maria Grazia Roncarolo; Pietro Luigi Poliani; Elisabetta Traggiai; Anna Villa

BACKGROUND Wiskott-Aldrich syndrome (WAS) is an X-linked primary immunodeficiency characterized by thrombocytopenia, eczema, infections, autoimmunity, and lymphomas. Transplantation of hematopoietic stem cells from HLA-identical donors is curative, but it is not available to all patients. We have developed a gene therapy (GT) approach for WAS by using a lentiviral vector encoding for human WAS promoter/cDNA (w1.6W) and demonstrated its preclinical efficacy and safety. OBJECTIVE To evaluate B-cell reconstitution and correction of B-cell phenotype in GT-treated mice. METHODS We transplanted Was(-/-) mice sublethally irradiated (700 rads) with lineage marker-depleted bone marrow wild-type cells, Was(-/-) cells untransduced or transduced with the w1.6W lentiviral vector and analyzed B-cell reconstitution in bone marrow, spleen, and peritoneum. RESULTS Here we show that WAS protein(+) B cells were present in central and peripheral B-cell compartments from GT-treated mice and displayed the strongest selective advantage in the splenic marginal zone and peritoneal B1 cell subsets. After GT, splenic architecture was improved and B-cell functions were restored, as demonstrated by the improved antibody response to pneumococcal antigens and the reduction of serum IgG autoantibodies. CONCLUSION WAS GT leads to improvement of B-cell functions, even in the presence of a mixed chimerism, further validating the clinical application of the w1.6W lentiviral vector.


The Journal of Allergy and Clinical Immunology | 2015

Single amino acid charge switch defines clinically distinct proline-serine-threonine phosphatase-interacting protein 1 (PSTPIP1)–associated inflammatory diseases

Dirk Holzinger; Selina Kathleen Fassl; Wilco de Jager; Peter Lohse; Ute F. Röhrig; Marco Gattorno; Alessia Omenetti; Sabrina Chiesa; Francesca Schena; Judith Austermann; Thomas Vogl; Douglas B. Kuhns; Steven M. Holland; Carlos Rodríguez-Gallego; Ricardo López-Almaraz; Juan I. Aróstegui; Elena Colino; Rosa Roldan; Smaragdi Fessatou; Bertrand Isidor; Sylvaine Poignant; Koichi Ito; Hans Joerg Epple; Jonathan A. Bernstein; Michael Jeng; Jennifer Frankovich; Geraldina Lionetti; Joseph A. Church; Peck Y. Ong; Mona LaPlant

BACKGROUND Hyperzincemia and hypercalprotectinemia (Hz/Hc) is a distinct autoinflammatory entity involving extremely high serum concentrations of the proinflammatory alarmin myeloid-related protein (MRP) 8/14 (S100A8/S100A9 and calprotectin). OBJECTIVE We sought to characterize the genetic cause and clinical spectrum of Hz/Hc. METHODS Proline-serine-threonine phosphatase-interacting protein 1 (PSTPIP1) gene sequencing was performed in 14 patients with Hz/Hc, and their clinical phenotype was compared with that of 11 patients with pyogenic arthritis, pyoderma gangrenosum, and acne (PAPA) syndrome. PSTPIP1-pyrin interactions were analyzed by means of immunoprecipitation and Western blotting. A structural model of the PSTPIP1 dimer was generated. Cytokine profiles were analyzed by using the multiplex immunoassay, and MRP8/14 serum concentrations were analyzed by using an ELISA. RESULTS Thirteen patients were heterozygous for a missense mutation in the PSTPIP1 gene, resulting in a p.E250K mutation, and 1 carried a mutation resulting in p.E257K. Both mutations substantially alter the electrostatic potential of the PSTPIP1 dimer model in a region critical for protein-protein interaction. Patients with Hz/Hc have extremely high MRP8/14 concentrations (2045 ± 1300 μg/mL) compared with those with PAPA syndrome (116 ± 74 μg/mL) and have a distinct clinical phenotype. A specific cytokine profile is associated with Hz/Hc. Hz/Hc mutations altered protein binding of PSTPIP1, increasing interaction with pyrin through phosphorylation of PSTPIP1. CONCLUSION Mutations resulting in charge reversal in the y-domain of PSTPIP1 (E→K) and increased interaction with pyrin cause a distinct autoinflammatory disorder defined by clinical and biochemical features not found in patients with PAPA syndrome, indicating a unique genotype-phenotype correlation for mutations in the PSTPIP1 gene. This is the first inborn autoinflammatory syndrome in which inflammation is driven by uncontrolled release of members of the alarmin family.


Annals of the Rheumatic Diseases | 2017

ADA2 deficiency (DADA2) as an unrecognised cause of early onset polyarteritis nodosa and stroke: a multicentre national study

Roberta Caorsi; Federica Penco; Alice Grossi; Antonella Insalaco; Alessia Omenetti; Maria Alessio; Giovanni Conti; Federico Marchetti; Paolo Picco; Alberto Tommasini; Silvana Martino; Clara Malattia; Romina Gallizi; Rosa Anna Podda; Annalisa Salis; Fernanda Falcini; Francesca Schena; Francesca Garbarino; Alessia Morreale; Manuela Pardeo; Claudia Ventrici; Chiara Passarelli; Qing Zhou; Mariasavina Severino; Carlo Gandolfo; Gianluca Damonte; Alberto Martini; Angelo Ravelli; Ivona Aksentijevich; Isabella Ceccherini

Objectives To analyse the prevalence of CECR1 mutations in patients diagnosed with early onset livedo reticularis and/or haemorrhagic/ischaemic strokes in the context of inflammation or polyarteritis nodosa (PAN). Forty-eight patients from 43 families were included in the study. Methods Direct sequencing of CECR1 was performed by Sanger analysis. Adenosine deaminase 2 (ADA2) enzymatic activity was analysed in monocyte isolated from patients and healthy controls incubated with adenosine and with or without an ADA1 inhibitor. Results Biallelic homozygous or compound heterozygous CECR1 mutations were detected in 15/48 patients. A heterozygous disease-associated mutation (p.G47V) was observed in two affected brothers. The mean age of onset of the genetically positive patients was 24 months (6 months to 7 years). Ten patients displayed one or more cerebral strokes during their disease course. Low immunoglobulin levels were detected in six patients. Thalidomide and anti-TNF (tumour necrosis factor) blockers were the most effective drugs. Patients without CECR1 mutations had a later age at disease onset, a lower prevalence of neurological and skin manifestations; one of these patients displayed all the clinical features of adenosine deaminase 2deficiency (DADA2) and a defective enzymatic activity suggesting the presence of a missed mutation or a synthesis defect. Conclusions DADA2 accounts for paediatric patients diagnosed with PAN-like disease and strokes and might explain an unrecognised condition in patients followed by adult rheumatologist. Timely diagnosis and treatment with anti-TNF agents are crucial for the prevention of severe complications of the disease. Functional assay to measure ADA2 activity should complement genetic testing in patients with non-confirming genotypes.


Journal of Bone and Mineral Research | 2012

Osteopetrosis rescue upon RANKL administration to Rankl−/− mice: A new therapy for human RANKL-dependent ARO

Nadia Lo Iacono; Harry C. Blair; Pietro Luigi Poliani; Veronica Marrella; Francesca Ficara; Barbara Cassani; Fabio Facchetti; Elena Fontana; Matteo M Guerrini; Elisabetta Traggiai; Francesca Schena; Marianna Paulis; Stefano Mantero; Antonio Inforzato; Serenella Valaperta; Alessandra Pangrazio; Laura Crisafulli; Virginia Maina; Paul J. Kostenuik; Paolo Vezzoni; Anna Villa; Cristina Sobacchi

In the last decades the molecular basis of monogenic diseases has been largely unraveled, although their treatment has often remained unsatisfactory. Autosomal recessive osteopetrosis (ARO) belongs to the small group of genetic diseases that are usually treated with hematopoietic stem cell transplantation (HSCT). However, this approach is not effective in the recently identified form carrying mutations in the receptor activator of NF‐κB ligand (RANKL) gene. In this subset, therapy replacement approach based on RANKL delivery has a strong rationale. Here we demonstrate that the systematic administration of RANKL for 1 month to Rankl−/− mice, which closely resemble the human disease, significantly improves the bone phenotype and has beneficial effects on bone marrow, spleen and thymus; major adverse effects arise only when mice are clearly overtreated. Overall, we provide evidence that the pharmacological administration of RANKL represents the appropriate treatment option for RANKL‐deficient ARO patients, to be validated in a pilot clinical trial.


Blood | 2012

Anti-CD3ε mAb improves thymic architecture and prevents autoimmune manifestations in a mouse model of Omenn syndrome: therapeutic implications.

Veronica Marrella; Pietro Luigi Poliani; Elena Fontana; Anna Casati; Virginia Maina; Barbara Cassani; Francesca Ficara; Manuela Cominelli; Francesca Schena; Marianna Paulis; Elisabetta Traggiai; Paolo Vezzoni; Fabio Grassi; Anna Villa

Omenn syndrome (OS) is an atypical primary immunodeficiency characterized by severe autoimmunity because of activated T cells infiltrating target organs. The impaired recombinase activity in OS severely affects expression of the pre-T-cell receptor complex in immature thymocytes, which is crucial for an efficient development of the thymic epithelial component. Anti-CD3ε monoclonal antibody (mAb) treatment in RAG2(-/-) mice was previously shown to mimic pre-TCR signaling promoting thymic expansion. Here we show the effect of anti-CD3ε mAb administration in the RAG2(R229Q) mouse model, which closely recapitulates human OS. These animals, in spite of the inability to induce the autoimmune regulator, displayed a significant amelioration in thymic epithelial compartment and an important reduction of peripheral T-cell activation and tissue infiltration. Furthermore, by injecting a high number of RAG2(R229Q) progenitors into RAG2(-/-) animals previously conditioned with anti-CD3ε mAb, we detected autoimmune regulator expression together with the absence of peripheral immunopathology. These observations indicate that improving epithelial thymic function might ameliorate the detrimental behavior of the cell-autonomous RAG defect. Our data provide important therapeutic proof of concept for future clinical applications of anti-CD3ε mAb treatment in severe combined immunodeficiency forms characterized by poor thymus function and autoimmunity.


Stem Cells | 2017

Murine Rankl‐/‐ Mesenchymal Stromal Cells Display an Osteogenic Differentiation Defect Improved by a RANKL‐expressing Lentiviral Vector

Francesca Schena; Ciro Menale; Emanuela Caci; Lorenzo Diomede; Eleonora Palagano; Camilla Recordati; Monica Sandri; Anna Tampieri; Ileana Bortolomai; Valentina Capo; Claudia Pastorino; Arinna Bertoni; Marco Gattorno; Alberto Martini; Anna Villa; Elisabetta Traggiai; Cristina Sobacchi

Autosomal recessive osteopetrosis (ARO) is a severe bone disease characterized by increased bone density due to impairment in osteoclast resorptive function or differentiation. Hematopoietic stem cell transplantation is the only available treatment; however, this therapy is not effective in RANKL‐dependent ARO, since in bone this gene is mainly expressed by cells of mesenchymal origin. Of note, whether lack of RANKL production might cause a defect also in the bone marrow (BM) stromal compartment, possibly contributing to the pathology, is unknown. To verify this possibility, we generated and characterized BM mesenchymal stromal cell (BM‐MSC) lines from wild type and Rankl−/− mice, and found that Rankl−/− BM‐MSCs displayed reduced clonogenicity and osteogenic capacity. The differentiation defect was significantly improved by lentiviral transduction of Rankl−/− BM‐MSCs with a vector stably expressing human soluble RANKL (hsRANKL). Expression of Rankl receptor, Rank, on the cytoplasmic membrane of BM‐MSCs pointed to the existence of an autocrine loop possibly activated by the secreted cytokine. Based on the close resemblance of RANKL‐defective osteopetrosis in humans and mice, we expect that our results are also relevant for RANKL‐dependent ARO patients. Data obtained in vitro after transduction with a lentiviral vector expressing hsRANKL would suggest that restoration of RANKL production might not only rescue the defective osteoclastogenesis of this ARO form, but also improve a less obvious defect in the osteoblast lineage, thus possibly achieving higher benefit for the patients, when the approach is translated to clinics. Stem Cells 2017;35:1365–1377

Collaboration


Dive into the Francesca Schena's collaboration.

Top Co-Authors

Avatar

Anna Villa

National Research Council

View shared research outputs
Top Co-Authors

Avatar

Alberto Martini

Istituto Giannina Gaslini

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Gattorno

Istituto Giannina Gaslini

View shared research outputs
Top Co-Authors

Avatar

Ciro Menale

National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge