Network


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

Hotspot


Dive into the research topics where Valentina Pettirossi is active.

Publication


Featured researches published by Valentina Pettirossi.


The New England Journal of Medicine | 2011

BRAF Mutations in Hairy-Cell Leukemia

Enrico Tiacci; Vladimir Trifonov; Gianluca Schiavoni; Antony B. Holmes; Wolfgang Kern; Maria Paola Martelli; Alessandra Pucciarini; Barbara Bigerna; Roberta Pacini; Victoria A. Wells; Paolo Sportoletti; Valentina Pettirossi; Roberta Mannucci; Oliver Elliott; Arcangelo Liso; Achille Ambrosetti; Alessandro Pulsoni; Francesco Forconi; Livio Trentin; Gianpietro Semenzato; Giorgio Inghirami; Monia Capponi; Francesco Di Raimondo; Caterina Patti; Luca Arcaini; Pellegrino Musto; Stefano Pileri; Claudia Haferlach; Susanne Schnittger; Giovanni Pizzolo

BACKGROUND Hairy-cell leukemia (HCL) is a well-defined clinicopathological entity whose underlying genetic lesion is still obscure. METHODS We searched for HCL-associated mutations by performing massively parallel sequencing of the whole exome of leukemic and matched normal cells purified from the peripheral blood of an index patient with HCL. Findings were validated by Sanger sequencing in 47 additional patients with HCL. RESULTS Whole-exome sequencing identified five missense somatic clonal mutations that were confirmed on Sanger sequencing, including a heterozygous mutation in BRAF that results in the BRAF V600E variant protein. Since BRAF V600E is oncogenic in other tumors, further analyses were focused on this genetic lesion. The same BRAF mutation was noted in all the other 47 patients with HCL who were evaluated by means of Sanger sequencing. None of the 195 patients with other peripheral B-cell lymphomas or leukemias who were evaluated carried the BRAF V600E variant, including 38 patients with splenic marginal-zone lymphomas or unclassifiable splenic lymphomas or leukemias. In immunohistologic and Western blot studies, HCL cells expressed phosphorylated MEK and ERK (the downstream targets of the BRAF kinase), indicating a constitutive activation of the RAF-MEK-ERK mitogen-activated protein kinase pathway in HCL. In vitro incubation of BRAF-mutated primary leukemic hairy cells from 5 patients with PLX-4720, a specific inhibitor of active BRAF, led to a marked decrease in phosphorylated ERK and MEK. CONCLUSIONS; The BRAF V600E mutation was present in all patients with HCL who were evaluated. This finding may have implications for the pathogenesis, diagnosis, and targeted therapy of HCL. (Funded by Associazione Italiana per la Ricerca sul Cancro and others.).


American Journal of Pathology | 2009

EML4-ALK Rearrangement in Non-Small Cell Lung Cancer and Non-Tumor Lung Tissues

Maria Paola Martelli; Gabriella Sozzi; Luis Hernández; Valentina Pettirossi; Alba Navarro; Davide Conte; Patrizia Gasparini; Federica Perrone; Piergiorgio Modena; Ugo Pastorino; Antonino Carbone; Alessandra Fabbri; Angelo Sidoni; Shigeo Nakamura; Marcello Gambacorta; Pedro L. Fernández; José Ramírez; John K.C. Chan; Walter Franco Grigioni; Elias Campo; Stefano Pileri; Brunangelo Falini

A fusion gene, echinoderm microtubule associated protein like 4-anaplastic lymphoma kinase (EML4-ALK), with transforming activity has recently been identified in a subset of non-small cell lung cancer (NSCLC), but its pathogenetic, diagnostic, and therapeutic roles remain unclear. Both frequency and type of EML4-ALK transcripts were investigated by reverse transcription PCR in 120 frozen NSCLC specimens from Italy and Spain; non-neoplastic lung tissues taken far from the tumor were used as controls. In cases carrying the fusion transcript, we determined EML4-ALK gene and protein levels using fluorescence in situ hybridization, Western blotting, and immunoprecipitation. We also analyzed ALK protein levels in paraffin samples from 662 NSCLC specimens, including the 120 cases investigated in the molecular studies. EML4-ALK transcripts (variants 1 and 3) were detected in 9 of 120 NSCLC samples but were not specific for NSCLC since they were also found in non-cancerous lung tissues taken far from the tumor. Notably, no transcripts were detected in matching tumor samples from these patients. Fluorescence in situ hybridization analysis of cases expressing EML4-ALK transcripts showed that only a minority of cells harbored the EML4-ALK gene. None of these cases was found to express the EML4-ALK protein as examined by immunohistochemistry, Western blotting, and immunoprecipitation. The EML4-ALK transcript cannot be regarded as a specific diagnostic tool for NSCLC. Our results show therefore that the causal role and value of EML4-ALK as a therapeutic target remain to be defined.


Leukemia | 2007

Cytoplasmic mutated nucleophosmin (NPM) defines the molecular status of a significant fraction of myeloid sarcomas.

Brunangelo Falini; D Lenze; Robert P. Hasserjian; Sarah E. Coupland; D Jaehne; Chad P. Soupir; Arcangelo Liso; M P Martelli; N. Bolli; Francesco Bacci; Valentina Pettirossi; Antonella Santucci; M F Martelli; Stefano Pileri; Harald Stein

Cytoplasmic mutated nucleophosmin (NPM) defines the molecular status of a significant fraction of myeloid sarcomas


Blood | 2010

CD34+ cells from AML with mutated NPM1 harbor cytoplasmic mutated nucleophosmin and generate leukemia in immunocompromised mice

Maria Paola Martelli; Valentina Pettirossi; Christian Thiede; Elisabetta Bonifacio; Federica Mezzasoma; Debora Cecchini; Roberta Pacini; Alessia Tabarrini; Raffaella Ciurnelli; Ilaria Gionfriddo; Nicla Manes; Roberta Rossi; Linda Giunchi; Lorenzo Brunetti; Marica Gemei; Mario Delia; Giorgina Specchia; Arcangelo Liso; Franca Falzetti; Luigi Del Vecchio; Massimo F. Martelli; Brunangelo Falini

Acute myeloid leukemia (AML) with mutated NPM1 shows distinctive biologic and clinical features, including absent/low CD34 expression, the significance of which remains unclear. Therefore, we analyzed CD34(+) cells from 41 NPM1-mutated AML. At flow cytometry, 31 of 41 samples contained less than 10% cells showing low intensity CD34 positivity and variable expression of CD38. Mutational analysis and/or Western blotting of purified CD34(+) cells from 17 patients revealed NPM1-mutated gene and/or protein in all. Immunohistochemistry of trephine bone marrow biopsies and/or flow cytometry proved CD34(+) leukemia cells from NPM1-mutated AML had aberrant nucleophosmin expression in cytoplasm. NPM1-mutated gene and/or protein was also confirmed in a CD34(+) subfraction exhibiting the phenotype (CD34(+)/CD38(-)/CD123(+)/CD33(+)/CD90(-)) of leukemic stem cells. When transplanted into immunocompromised mice, CD34(+) cells generated a leukemia recapitulating, both morphologically and immunohistochemically (aberrant cytoplasmic nucleophosmin, CD34 negativity), the original patients disease. These results indicate that the CD34(+) fraction in NPM1-mutated AML belongs to the leukemic clone and contains NPM1-mutated cells exhibiting properties typical of leukemia-initiating cells. CD34(-) cells from few cases (2/15) also showed significant leukemia-initiating cell potential in immunocompromised mice. This study provides further evidence that NPM1 mutation is a founder genetic lesion and has potential implications for the cell-of-origin and targeted therapy of NPM1-mutated AML.


Blood Reviews | 2011

Acute myeloid leukemia with mutated nucleophosmin (NPM1): Any hope for a targeted therapy?

Brunangelo Falini; Ilaria Gionfriddo; Federica Cecchetti; Stelvio Ballanti; Valentina Pettirossi; Maria Paola Martelli

Acute myeloid leukemia (AML) carrying nucleophosmin (NPM1) mutations displays distinct molecular and clinical-pathological features that led to its inclusion as provisional entity in 2008 WHO classification of myeloid neoplasms. Since NPM1 mutations behave as a founder genetic lesion in AML, they could be an attractive target for therapeutic intervention. Here, we discuss the potential for developing targeted therapies for NPM1-mutated AML with focus on: (i) interfering with the abnormal traffic of the NPM1 leukemic mutant, i.e., its cytoplasmic dislocation; (ii) disrupting the nucleolar structure/function by interfering with residual wild-type nucleophosmin and other nucleolar components acting as hub proteins; and (iii) evaluating the activity of epigenetic drugs (e.g., 5-azacytidine) or agents acting on differentiation and apoptosis. As quantitative assessment of NPM1 mutated transcript copies now provides the means to measure minimal residual disease, we also discuss the potential for intervening in NPM1-mutated AML before overt hematological relapse occurs (so-called pre-emptive therapy).


Haematologica | 2013

Constant activation of the RAF-MEK-ERK pathway as a diagnostic and therapeutic target in hairy cell leukemia

Enrico Tiacci; Gianluca Schiavoni; Maria Paola Martelli; Emanuela Boveri; Roberta Pacini; Alessia Tabarrini; Silvia Zibellini; Alessia Santi; Valentina Pettirossi; Elisabetta Fortini; Stefano Ascani; Luca Arcaini; Giorgio Inghirami; Marco Paulli; Brunangelo Falini

The BRAF-V600E mutation defines genetically hairy cell leukemia among B-cell leukemias and lymphomas. In solid tumors, BRAF-V600E is known to aberrantly activate the oncogenic MEK-ERK pathway, and targeted BRAF and/or MEK inhibitors have shown remarkable efficacy in clinical trials in melanoma patients. However, the MEK-ERK pathway status in hairy cell leukemia has not been thoroughly investigated. We assessed phospho-ERK expression in 37 patients with hairy cell leukemia and 44 patients with neoplasms mimicking hairy cell leukemia (40 splenic marginal zone lymphoma, 2 hairy cell leukemia-variant and 2 splenic lymphoma/leukemia unclassifiable) using immunohistochemistry on routine biopsies and/or Western blotting on purified leukemic cells, and correlated the phospho-ERK status with the BRAF-V600E mutation status. Besides confirming the constant presence of BRAF-V600E in all patients with hairy cell leukemia, we observed ubiquitous phospho-ERK expression in this malignancy. Conversely, all 44 cases with neoplasms mimicking hairy cell leukemia were devoid of BRAF-V600E and none expressed phospho-ERK. Furthermore, the two exceptionally rare cases of non-hairy cell leukemia unclassifiable chronic B-cell neoplasms previously reported to be BRAF-V600E+ on allele-specific polymerase chain reaction lacked phospho-ERK expression as well, suggesting the presence of the mutation in only a small part of the leukemic clone in these cases. In conclusion, our findings support the use of phospho-ERK immunohistochemistry in the differential diagnosis between hairy cell leukemia and its mimics, and establish the MEK-ERK pathway as a rational therapeutic target in this malignancy.


Leukemia | 2009

A dose-dependent tug of war involving the NPM1 leukaemic mutant, nucleophosmin, and ARF

N Bolli; M F De Marco; M P Martelli; Barbara Bigerna; Alessandra Pucciarini; R Rossi; Roberta Mannucci; N. Manes; Valentina Pettirossi; S A Pileri; Ildo Nicoletti; Brunangelo Falini

In acute myeloid leukaemia (AML), nucleophosmin-1 (NPM1) mutations create a nuclear export signal (NES) motif and disrupt tryptophans at NPM1 C-terminus, leading to nucleophosmin accumulation in leukaemic cell cytoplasm. We investigated how nucleophosmin NES motifs (two physiological and one created by the mutation) regulate traffic and interaction of mutated NPM1, NPM1wt and p14ARF. Nucleophosmin export into cytoplasm was maximum when the protein contained all three NES motifs, as naturally occurs in NPM1-mutated AML. The two physiological NES motifs mediated NPM1 homo/heterodimerization, influencing subcellular distribution of NPM1wt, mutated NPM1 and p14ARF in a ‘dose-dependent tug of war’ fashion. In transfected cells, excess doses of mutant NPM1 relocated completely NPM1wt (and p14ARF) from the nucleoli to the cytoplasm. This distribution pattern was also observed in a proportion of NPM1-mutated AML patients. In transfected cells, excess of NPM1wt (and p14ARF) relocated NPM1 mutant from the cytoplasm to the nucleoli. Notably, this distribution pattern was not observed in AML patients where the mutant was consistently cytoplasmic restricted. These findings reinforce the concept that NPM1 mutants are naturally selected for most efficient cytoplasmic export, pointing to this event as critical for leukaemogenesis. Moreover, they provide a rationale basis for designing small molecules acting at the interface between mutated NPM1 and other interacting proteins.


Leukemia | 2008

A western blot assay for detecting mutant nucleophosmin (NPM1) proteins in acute myeloid leukaemia

M P Martelli; Nicla Manes; Arcangelo Liso; Valentina Pettirossi; B. Verducci Galletti; Barbara Bigerna; Alessandra Pucciarini; M F De Marco; M. T. Pallotta; Niccolo Bolli; Marco Sborgia; F. Di Raimondo; F Fabbiano; Giovanna Meloni; G Specchia; M F Martelli

A western blot assay for detecting mutant nucleophosmin (NPM1) proteins in acute myeloid leukaemia


Leukemia | 2008

Absence of nucleophosmin leukaemic mutants in B and T cells from AML with NPM1 mutations: implications for the cell of origin of NPMc+ AML.

Maria Paola Martelli; Nicla Manes; Valentina Pettirossi; Arcangelo Liso; Roberta Pacini; Roberta Mannucci; Tiziana Zei; Niccolo Bolli; F. Di Raimondo; G Specchia; Ildo Nicoletti; Massimo F. Martelli; Brunangelo Falini

Absence of nucleophosmin leukaemic mutants in B and T cells from AML with NPM1 mutations: implications for the cell of origin of NPMc+ AML


Journal of Cell Science | 2005

HOPS: a novel cAMP-dependent shuttling protein involved in protein synthesis regulation

Maria Agnese Della Fazia; Marilena Castelli; Daniela Bartoli; Stefania Pieroni; Valentina Pettirossi; Danilo Piobbico; Mariapia Viola-Magni; Giuseppe Servillo

The liver has the ability to autonomously regulate growth and mass. Following partial hepatectomy, hormones, growth factors, cytokines and their coupled signal transduction pathways have been implicated in hepatocyte proliferation. To understand the mechanisms responsible for the proliferative response, we studied liver regeneration by characterization of novel genes that are activated in residual hepatocytes. A regenerating liver cDNA library screening was performed with cDNA-subtracted probes derived from regenerating and normal liver. Here, we describe the biology of Hops (for hepatocyte odd protein shuttling). HOPS is a novel shuttling protein that contains an ubiquitin-like domain, a putative NES and a proline-rich region. HOPS is rapidly exported from the nucleus and is overexpressed during liver regeneration. Evidence shows that cAMP governs HOPS export in hepatocytes of normal and regenerating liver and is mediated via CRM-1. We demonstrate that HOPS binds to elongation factor eEF-1A and interferes in protein synthesis. HOPS overexpression in H-35-hepatoma and 3T3-NIH cells strongly reduces proliferation.

Collaboration


Dive into the Valentina Pettirossi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge