Network


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

Hotspot


Dive into the research topics where Veronica Marrella is active.

Publication


Featured researches published by Veronica Marrella.


Journal of Clinical Investigation | 2005

AIRE deficiency in thymus of 2 patients with Omenn syndrome

Patrizia Cavadini; William Vermi; Fabio Facchetti; Stefania Fontana; Seiho Nagafuchi; Evelina Mazzolari; Anna Sediva; Veronica Marrella; Anna Villa; Alain Fischer; Luigi D. Notarangelo; Raffaele Badolato

Omenn syndrome is a severe primary immunodeficiency with putative autoimmune manifestations of the skin and gastrointestinal tract. The disease is caused by hypomorphic mutations in recombination-activating genes that impair but do not abolish the process of VDJ recombination, leading to the generation of autoreactive T cells with a highly restricted receptor repertoire. Loss of central tolerance in genetically determined autoimmune diseases, e.g., autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, is associated with defective expression by medullary thymic epithelial cells of AIRE, the transcription activator that induces thymic expression of tissue-specific antigens. Analysis of AIRE expression in the thymi of 2 Omenn syndrome patients and 1 SCID patient, by real-time RT-PCR and immunohistochemistry, demonstrated a profound reduction in the levels of AIRE mRNA and protein in patients as compared with a normal control subject. Lack of AIRE was associated with normal or even increased levels of keratin and lymphotoxin-beta receptor mRNAs, while mRNAs of the self-antigens insulin, cytochrome P450 1a2, and fatty acid-binding protein were undetectable in thymi from immunodeficiency patients. These results demonstrate that deficiency of AIRE expression is observed in severe immunodeficiencies characterized by abnormal T cell development and suggest that in Omenn syndrome, the few residual T cell clones that develop may escape negative selection and thereafter expand in the periphery, causing massive autoimmune reactions.


Journal of Clinical Investigation | 2007

A hypomorphic R229Q Rag2 mouse mutant recapitulates human Omenn syndrome

Veronica Marrella; Pietro Luigi Poliani; Anna Casati; Francesca Rucci; Laura Frascoli; Marie Lise Gougeon; Brigitte Lemercier; Marita Bosticardo; Maria Ravanini; Manuela Battaglia; Maria Grazia Roncarolo; Marina Cavazzana-Calvo; Fabio Facchetti; Luigi D. Notarangelo; Paolo Vezzoni; Fabio Grassi; Anna Villa

Rag enzymes are the main players in V(D)J recombination, the process responsible for rearrangement of TCR and Ig genes. Hypomorphic Rag mutations in humans, which maintain partial V(D)J activity, cause a peculiar SCID associated with autoimmune-like manifestations, Omenn syndrome (OS). Although a deficient ability to sustain thymopoiesis and to produce a diverse T and B cell repertoire explains the increased susceptibility to severe infections, the molecular and cellular mechanisms underlying the spectrum of clinical and immunological features of OS remain poorly defined. In order to better define the molecular and cellular pathophysiology of OS, we generated a knockin murine model carrying the Rag2 R229Q mutation previously described in several patients with OS and leaky forms of SCID. These Rag2(R229Q/R229Q) mice showed oligoclonal T cells, absence of circulating B cells, and peripheral eosinophilia. In addition, activated T cells infiltrated gut and skin, causing diarrhea, alopecia, and, in some cases, severe erythrodermia. These findings were associated with reduced thymic expression of Aire and markedly reduced numbers of naturally occurring Tregs and NKT lymphocytes. In conclusion, Rag2(R229Q/R229Q) mice mimicked most symptoms of human OS; our findings support the notion that impaired immune tolerance and defective immune regulation are involved in the pathophysiology of OS.


The Journal of Allergy and Clinical Immunology | 2010

Defect of regulatory T cells in patients with Omenn syndrome

Barbara Cassani; Pietro Luigi Poliani; Daniele Moratto; Cristina Sobacchi; Veronica Marrella; Laura Imperatori; Donatella Vairo; Alessandro Plebani; Silvia Giliani; Paolo Vezzoni; Fabio Facchetti; Fulvio Porta; Luigi D. Notarangelo; Anna Villa; Raffaele Badolato

BACKGROUND Omenn syndrome (OS) is an autosomal-recessive disorder characterized by severe immunodeficiency and T-cell-mediated autoimmunity. The disease is caused by hypomorphic mutations in recombination-activating genes that hamper the process of Variable (V) Diversity (D) Joining (J) recombination, leading to the generation of autoreactive T cells. We have previously shown that in OS the expression of autoimmune regulator, a key factor governing central tolerance, is markedly reduced. OBJECTIVE Here, we have addressed the role of peripheral tolerance in the disease pathogenesis. METHODS We have analyzed forkhead box protein P3 (FOXP3) expression in peripheral blood T cells of 4 patients with OS and in lymphoid organs of 8 patients with OS and have tested the suppressive activity of sorted CD4(+) CD25(high) peripheral blood T cells in 2 of these patients. RESULTS We have observed that CD4(+)CD25(high)T cells isolated ex vivo from patients with OS failed to suppress proliferation of autologous or allogenic CD4(+) responder T cells. Moreover, despite individual variability in the fraction of circulating FOXP3(+) CD4 cells in patients with OS, the immunohistochemical analysis of FOXP3 expression in lymph nodes and thymus of patients with OS demonstrated a severe reduction of this cell subset compared with control tissues. CONCLUSION Overall, these results suggest a defect of regulatory T cells in OS leading to a breakdown of peripheral tolerance, which may actively concur to the development of autoimmune manifestations in the disease.


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.


Journal of Bone and Mineral Research | 2012

RANK-Dependent Autosomal Recessive Osteopetrosis: Characterization of Five New Cases With Novel Mutations

Alessandra Pangrazio; Barbara Cassani; Matteo M Guerrini; Julie C. Crockett; Veronica Marrella; Luca Zammataro; Dario Strina; Ansgar Schulz; Claire Schlack; Uwe Kornak; David Mellis; Angela Duthie; Miep H. Helfrich; Anne Durandy; Despina Moshous; Ashok Vellodi; Robert Chiesa; Paul Veys; Nadia Lo Iacono; Paolo Vezzoni; Alain Fischer; Anna Villa; Cristina Sobacchi

Autosomal recessive osteopetrosis (ARO) is a genetically heterogeneous disorder attributed to reduced bone resorption by osteoclasts. Most human AROs are classified as osteoclast rich, but recently two subsets of osteoclast‐poor ARO have been recognized as caused by defects in either TNFSF11 or TNFRSF11A genes, coding the RANKL and RANK proteins, respectively. The RANKL/RANK axis drives osteoclast differentiation and also plays a role in the immune system. In fact, we have recently reported that mutations in the TNFRSF11A gene lead to osteoclast‐poor osteopetrosis associated with hypogammaglobulinemia. Here we present the characterization of five additional unpublished patients from four unrelated families in which we found five novel mutations in the TNFRSF11A gene, including two missense and two nonsense mutations and a single‐nucleotide insertion. Immunological investigation in three of them showed that the previously described defect in the B cell compartment was present only in some patients and that its severity seemed to increase with age and the progression of the disease. HSCT performed in all five patients almost completely cured the disease even when carried out in late infancy. Hypercalcemia was the most important posttransplant complication. Overall, our results further underline the heterogeneity of human ARO also deriving from the interplay between bone and the immune system, and highlight the prognostic and therapeutic implications of the molecular diagnosis.


European Journal of Immunology | 2005

Damaging-agent sensitivity of Artemis-deficient cell lines

Antonio Musio; Veronica Marrella; Cristina Sobacchi; Francesca Rucci; Laura Fariselli; Silvia Giliani; Gaetana Lanzi; Luigi D. Notarangelo; Domenico Delia; Roberto Colombo; Paolo Vezzoni; Anna Villa

Defects in repairing double‐strand breaks can lead to genome instability and tumorigenesis. In humans, most T–B– severe combined immunodeficiencies (SCID) have a defect in either the RAG1 or RAG2 gene, are not radiosensitive and do not show genome instability. On the contrary, a minority of T–B– SCID patients have abnormalities in the Artemis gene and are moderately radiosensitive. Artemis‐deficient cells are unable to process hairpin ends after RAG cleavage, but hairpin opening activity alone does not explain the moderate X‐ray sensitivity of Artemis‐deficient cells. We report here that, at variance with what has been described in mice, cell lines from Artemis–/– patients are moderately sensitive to mitomycin C and show only a low to moderate increase in genomic instability, both spontaneously and after exposure to ionizing radiations. There is some heterogeneity in the levels of DNA damage sensitivity and genome instability, which could in part be due to different effects of the specific mutation involved or to genetic background, which may not always represent null alleles. This data supports the hypothesis that, in addition to playing a role in hairpin opening during the V(D)J recombination process, Artemis is involved in the repair of a subset of DNA damage whose exact nature is still undefined.


Current Opinion in Allergy and Clinical Immunology | 2011

Omenn syndrome does not live by V(D)J recombination alone.

Veronica Marrella; Virginia Maina; Anna Villa

Purpose of review During the past decade, easy access to sequence analyses has allowed us to increase our understanding of the pathogenesis of severe combined immunodeficiencies. Here, we describe the expanding clinical and immunological spectrum associated with Omenn syndrome phenotype. In particular, we review the cellular and molecular mechanisms involved in the pathophysiology of ‘classical’ Omenn syndrome due to the recombination activating gene (RAG) defects and of a new subgroup of Omenn-like disorders. Recent findings Different types of mutations are associated with the Omenn phenotype characterized by skin erythroderma, oligoclonal-activated T cells and elevated IgE in the absence of circulating B cells. Extensive studies conducted over the last few years have allowed the definition of the ‘classical form’ of Omenn syndrome due to hypomorphic defects in genes involved in V(D)J recombination, mainly RAG genes, and ‘Omenn-like’ features associated with mutations in genes involved in the maturation steps of lymphoid cells other than V(D)J recombination. Moreover, an increasing number of diseases other than those due to V(D)J recombination defects develop Omenn signs. Summary Impaired but not abolished V(D)J recombination process leads to the generation of a few T cells which expand in the periphery, infiltrate target organs such as skin and gut, resulting in severe erythroderma and colitis, both typical signs of Omenn syndrome. Extensive molecular studies now demonstrate that genes other than V(D)J molecules have a role in the pathogenesis of this disease, supporting the evidence that ‘Omenn’ defines an inflammatory condition associated with various genetic defects.


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.


Trends in Immunology | 2008

Of Omenn and mice

Veronica Marrella; Pietro Luigi Poliani; Cristina Sobacchi; Fabio Grassi; Anna Villa

Omenn syndrome (OS) is a peculiar immunodeficiency in which profound T and B cell defects are associated with severe autoimmune manifestations. Although the molecular and biochemical bases of OS have been elucidated, the mechanisms leading to T cell infiltration of peripheral tissues such as skin and gut still remain unsolved. Two murine models with hypomorphic mutations in rag genes reproducing OS features and a murine model of lymphopenia-derived autoimmunity with similar immunopathology were recently described. The aim of this review is to integrate clues regarding the roles of impaired thymic development and lymphopenia into the pathogenesis of autoimmunity.

Collaboration


Dive into the Veronica Marrella's collaboration.

Top Co-Authors

Avatar

Anna Villa

National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paolo Vezzoni

National Research Council

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

Francesca Rucci

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge