Network


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

Hotspot


Dive into the research topics where Sabina Chiaretti is active.

Publication


Featured researches published by Sabina Chiaretti.


Cell | 2007

A Mammalian microRNA Expression Atlas Based on Small RNA Library Sequencing

Pablo Landgraf; Mirabela Rusu; Robert L. Sheridan; Alain Sewer; Nicola Iovino; Alexei A. Aravin; Sébastien Pfeffer; Amanda Rice; Alice O. Kamphorst; Markus Landthaler; Carolina Lin; Nicholas D. Socci; Leandro C. Hermida; Valerio Fulci; Sabina Chiaretti; Robin Foà; Julia Schliwka; Uta Fuchs; Astrid Novosel; Roman Ulrich Müller; Bernhard Schermer; Ute Bissels; Jason M. Inman; Quang Phan; Minchen Chien; David B. Weir; Ruchi Choksi; Gabriella De Vita; Daniela Frezzetti; Hans Ingo Trompeter

MicroRNAs (miRNAs) are small noncoding regulatory RNAs that reduce stability and/or translation of fully or partially sequence-complementary target mRNAs. In order to identify miRNAs and to assess their expression patterns, we sequenced over 250 small RNA libraries from 26 different organ systems and cell types of human and rodents that were enriched in neuronal as well as normal and malignant hematopoietic cells and tissues. We present expression profiles derived from clone count data and provide computational tools for their analysis. Unexpectedly, a relatively small set of miRNAs, many of which are ubiquitously expressed, account for most of the differences in miRNA profiles between cell lineages and tissues. This broad survey also provides detailed and accurate information about mature sequences, precursors, genome locations, maturation processes, inferred transcriptional units, and conservation patterns. We also propose a subclassification scheme for miRNAs for assisting future experimental and computational functional analyses.


Journal of Clinical Oncology | 2010

Clinical Utility of Microarray-Based Gene Expression Profiling in the Diagnosis and Subclassification of Leukemia: Report From the International Microarray Innovations in Leukemia Study Group

Torsten Haferlach; Alexander Kohlmann; Lothar Wieczorek; Giuseppe Basso; Geertruy te Kronnie; Marie C. Béné; John De Vos; Jesús Hernández; Wolf K. Hofmann; Ken I. Mills; Amanda F. Gilkes; Sabina Chiaretti; Sheila A. Shurtleff; Thomas J. Kipps; Laura Z. Rassenti; Allen Eng Juh Yeoh; Peter Papenhausen; Wei-min Liu; P. Mickey Williams; Robin Foà

PURPOSE The Microarray Innovations in Leukemia study assessed the clinical utility of gene expression profiling as a single test to subtype leukemias into conventional categories of myeloid and lymphoid malignancies. METHODS The investigation was performed in 11 laboratories across three continents and included 3,334 patients. An exploratory retrospective stage I study was designed for biomarker discovery and generated whole-genome expression profiles from 2,143 patients with leukemias and myelodysplastic syndromes. The gene expression profiling-based diagnostic accuracy was further validated in a prospective second study stage of an independent cohort of 1,191 patients. RESULTS On the basis of 2,096 samples, the stage I study achieved 92.2% classification accuracy for all 18 distinct classes investigated (median specificity of 99.7%). In a second cohort of 1,152 prospectively collected patients, a classification scheme reached 95.6% median sensitivity and 99.8% median specificity for 14 standard subtypes of acute leukemia (eight acute lymphoblastic leukemia and six acute myeloid leukemia classes, n = 693). In 29 (57%) of 51 discrepant cases, the microarray results had outperformed routine diagnostic methods. CONCLUSION Gene expression profiling is a robust technology for the diagnosis of hematologic malignancies with high accuracy. It may complement current diagnostic algorithms and could offer a reliable platform for patients who lack access to todays state-of-the-art diagnostic work-up. Our comprehensive gene expression data set will be submitted to the public domain to foster research focusing on the molecular understanding of leukemias.


Journal of Experimental Medicine | 2008

Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia

Elisabetta Flex; Valentina Petrangeli; Lorenzo Stella; Sabina Chiaretti; Tekla Hornakova; Laurent Knoops; Cristina Ariola; Valentina Fodale; Emmanuelle Clappier; Francesca Paoloni; Simone Martinelli; Alessandra Fragale; Massimo Sanchez; Simona Tavolaro; Monica Messina; Giovanni Cazzaniga; Andrea Camera; Giovanni Pizzolo; Assunta Tornesello; Marco Vignetti; Angela Battistini; Hélène Cavé; Bruce D. Gelb; Jean-Christophe Renauld; Andrea Biondi; Stefan N. Constantinescu; Robin Foà; Marco Tartaglia

Aberrant signal transduction contributes substantially to leukemogenesis. The Janus kinase 1 (JAK1) gene encodes a cytoplasmic tyrosine kinase that noncovalently associates with a variety of cytokine receptors and plays a nonredundant role in lymphoid cell precursor proliferation, survival, and differentiation. We report that somatic mutations in JAK1 occur in individuals with acute lymphoblastic leukemia (ALL). JAK1 mutations were more prevalent among adult subjects with the T cell precursor ALL, where they accounted for 18% of cases, and were associated with advanced age at diagnosis, poor response to therapy, and overall prognosis. All mutations were missense, and some were predicted to destabilize interdomain interactions controlling the activity of the kinase. Three mutations that were studied promoted JAK1 gain of function and conferred interleukin (IL)-3–independent growth in Ba/F3 cells and/or IL-9–independent resistance to dexamethasone-induced apoptosis in T cell lymphoma BW5147 cells. Such effects were associated with variably enhanced activation of multiple downstream signaling pathways. Leukemic cells with mutated JAK1 alleles shared a gene expression signature characterized by transcriptional up-regulation of genes positively controlled by JAK signaling. Our findings implicate dysregulated JAK1 function in ALL, particularly of T cell origin, and point to this kinase as a target for the development of novel antileukemic drugs.


Blood | 2011

Mutations of the SF3B1 splicing factor in chronic lymphocytic leukemia: association with progression and fludarabine-refractoriness

Davide Rossi; Alessio Bruscaggin; Valeria Spina; Silvia Rasi; Hossein Khiabanian; Monica Messina; Marco Fangazio; Tiziana Vaisitti; Sara Monti; Sabina Chiaretti; Anna Guarini; Ilaria Del Giudice; Michaela Cerri; Stefania Cresta; Clara Deambrogi; Ernesto Gargiulo; Valter Gattei; Francesco Forconi; Francesco Bertoni; Silvia Deaglio; Raul Rabadan; Laura Pasqualucci; Robin Foà; Riccardo Dalla-Favera; Gianluca Gaidano

The genetic lesions identified in chronic lymphocytic leukemia (CLL) do not entirely recapitulate the disease pathogenesis and the development of serious complications, such as chemorefractoriness. While investigating the coding genome of fludarabine-refractory CLL, we observed that mutations of SF3B1, encoding a splicing factor and representing a critical component of the cell spliceosome, were recurrent in 10 of 59 (17%) fludarabine-refractory cases, with a frequency significantly greater than that observed in a consecutive CLL cohort sampled at diagnosis (17/301, 5%; P = .002). Mutations were somatically acquired, were generally represented by missense nucleotide changes, clustered in selected HEAT repeats of the SF3B1 protein, recurrently targeted 3 hotspots (codons 662, 666, and 700), and were predictive of a poor prognosis. In fludarabine-refractory CLL, SF3B1 mutations and TP53 disruption distributed in a mutually exclusive fashion (P = .046). The identification of SF3B1 mutations points to splicing regulation as a novel pathogenetic mechanism of potential clinical relevance in CLL.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia

Akinori Yoda; Yuka Yoda; Sabina Chiaretti; Michal Bar-Natan; Kartik Mani; Scott J. Rodig; Nathan West; Yun Xiao; Jennifer R. Brown; Constantine S. Mitsiades; Martin Sattler; Jeffrey L. Kutok; Daniel J. DeAngelo; Martha Wadleigh; Alfonso Piciocchi; Paola Dal Cin; James E. Bradner; James D. Griffin; Kenneth C. Anderson; Richard Stone; Jerome Ritz; Robin Foà; David A. Frank; David M. Weinstock

The prognosis for adults with precursor B-cell acute lymphoblastic leukemia (B-ALL) remains poor, in part from a lack of therapeutic targets. We identified the type I cytokine receptor subunit CRLF2 in a functional screen for B-ALL–derived mRNA transcripts that can substitute for IL3 signaling. We demonstrate that CRLF2 is overexpressed in approximately 15% of adult and high-risk pediatric B-ALL that lack MLL, TCF3, TEL, and BCR/ABL rearrangements, but not in B-ALL with these rearrangements or other lymphoid malignancies. CRLF2 overexpression can result from translocation with the IGH locus or intrachromosomal deletion and is associated with poor outcome. CRLF2 overexpressing B-ALLs share a transcriptional signature that significantly overlaps with a BCR/ABL signature, and is enriched for genes involved in cytokine receptor and JAK-STAT signaling. In a subset of cases, CRLF2 harbors a Phe232Cys gain-of-function mutation that promotes constitutive dimerization and cytokine independent growth. A mutually exclusive subset harbors activating mutations in JAK2. In fact, all 22 B-ALLs with mutant JAK2 that we analyzed overexpress CRLF2, distinguishing CRLF2 as the key scaffold for mutant JAK2 signaling in B-ALL. Expression of WT CRLF2 with mutant JAK2 also promotes cytokine independent growth that, unlike CRLF2 Phe232Cys or ligand-induced signaling by WT CRLF2, is accompanied by JAK2 phosphorylation. Finally, cells dependent on CRLF2 signaling are sensitive to small molecule inhibitors of either JAKs or protein kinase C family kinases. Together, these findings implicate CRLF2 as an important factor in B-ALL with diagnostic, prognostic, and therapeutic implications.


Blood | 2012

Disruption of BIRC3 associates with fludarabine chemorefractoriness in TP53 wild-type chronic lymphocytic leukemia

Davide Rossi; Marco Fangazio; Silvia Rasi; Tiziana Vaisitti; Sara Monti; Stefania Cresta; Sabina Chiaretti; Ilaria Del Giudice; Giulia Fabbri; Alessio Bruscaggin; Valeria Spina; Clara Deambrogi; Marilisa Marinelli; Rosella Famà; Mariangela Greco; Giulia Daniele; Francesco Forconi; Valter Gattei; Francesco Bertoni; Silvia Deaglio; Laura Pasqualucci; Anna Guarini; Riccardo Dalla-Favera; Robin Foà; Gianluca Gaidano

The genetic lesions identified to date do not fully recapitulate the molecular pathogenesis of chronic lymphocytic leukemia (CLL) and do not entirely explain the development of severe complications such as chemorefractoriness. In the present study, BIRC3, a negative regulator of noncanonical NF-κB signaling, was investigated in different CLL clinical phases. BIRC3 lesions were absent in monoclonal B-cell lymphocytosis (0 of 63) and were rare in CLL at diagnosis (13 of 306, 4%). Conversely, BIRC3 disruption selectively affected 12 of 49 (24%) fludarabine-refractory CLL cases by inactivating mutations and/or gene deletions that distributed in a mutually exclusive fashion with TP53 abnormalities. In contrast to fludarabine-refractory CLL, progressive but fludarabine-sensitive patients were consistently devoid of BIRC3 abnormalities, suggesting that BIRC3 genetic lesions associate specifically with a chemorefractory phenotype. By actuarial analysis in newly diagnosed CLL (n = 306), BIRC3 disruption identified patients with a poor outcome similar to that associated with TP53 abnormalities and exerted a prognostic role that was independent of widely accepted clinical and genetic risk factors. Consistent with the role of BIRC3 as a negative regulator of NF-κB, biochemical studies revealed the presence of constitutive noncanonical NF-κB activation in fludarabine-refractory CLL patients harboring molecular lesions of BIRC3. These data identify BIRC3 disruption as a recurrent genetic lesion of high-risk CLL devoid of TP53 abnormalities.


Blood | 2009

Identification and molecular characterization of recurrent genomic deletions on 7p12 in the IKZF1 gene in a large cohort of BCR-ABL1-positive acute lymphoblastic leukemia patients: on behalf of Gruppo Italiano Malattie Ematologiche dell'Adulto Acute Leukemia Working Party (GIMEMA AL WP)

Ilaria Iacobucci; Clelia Tiziana Storlazzi; Daniela Cilloni; Annalisa Lonetti; Emanuela Ottaviani; Simona Soverini; Annalisa Astolfi; Sabina Chiaretti; Antonella Vitale; Francesca Messa; Luciana Impera; Carmen Baldazzi; Pietro D'Addabbo; Cristina Papayannidis; Angelo Lonoce; Sabrina Colarossi; Marco Vignetti; Pier Paolo Piccaluga; Stefania Paolini; Domenico Russo; Fabrizio Pane; Giuseppe Saglio; Michele Baccarani; Robin Foà; Giovanni Martinelli

The BCR-ABL1 fusion gene defines the subgroup of acute lymphoblastic leukemia (ALL) with the worst clinical prognosis. To identify oncogenic lesions that combine with BCR-ABL1 to cause ALL, we used Affymetrix Genome-Wide Human SNP arrays (250K NspI and SNP 6.0), fluorescence in situ hybridization, and genomic polymerase chain reaction to study 106 cases of adult BCR-ABL1-positive ALL. The most frequent somatic copy number alteration was a focal deletion on 7p12 of IKZF1, which encodes the transcription factor Ikaros and was identified in 80 (75%) of 106 patients. Different patterns of deletions occurred, but the most frequent were those characterized by a loss of exons 4 through 7 (Delta4-7) and by removal of exons 2 through 7 (Delta2-7). A variable number of nucleotides (patient specific) were inserted at the conjunction and maintained with fidelity at the time of relapse. The extent of the Delta4-7 deletion correlated with the expression of a dominant-negative isoform with cytoplasmic localization and oncogenic activity, whereas the Delta2-7 deletion resulted in a transcript lacking the translation start site. The IKZF1 deletion also was identified in the progression of chronic myeloid leukemia to lymphoid blast crisis (66%) but never in myeloid blast crisis or chronic-phase chronic myeloid leukemia or in patients with acute myeloid leukemia. Known DNA sequences and structural features were mapped along the breakpoint cluster regions, including heptamer recombination signal sequences recognized by RAG enzymes during V(D)J recombination, suggesting that IKZF1 deletions could arise from aberrant RAG-mediated recombination.


Haematologica | 2012

NOTCH1 mutations in +12 chronic lymphocytic leukemia (CLL) confer an unfavorable prognosis, induce a distinctive transcriptional profiling and refine the intermediate prognosis of +12 CLL

Ilaria Del Giudice; Davide Rossi; Sabina Chiaretti; Marilisa Marinelli; Simona Tavolaro; Sara Gabrielli; Luca Laurenti; Roberto Marasca; Silvia Rasi; Marco Fangazio; Anna Guarini; Gianluca Gaidano; Robin Foà

Trisomy 12, the third most frequent chromosomal aberration in chronic lymphocytic leukemia (CLL), confers an intermediate prognosis. In our cohort of 104 untreated patients carrying +12, NOTCH1 mutations occurred in 24% of cases and were associated to unmutated IGHV genes (P=0.003) and +12 as a sole cytogenetic abnormality (P=0.008). NOTCH1 mutations in +12 CLL associated with an approximately 2.4 fold increase in the risk of death, a significant shortening of survival (P<0.01) and proved to be an independent predictor of survival in multivariate analysis. Analogous to +12 CLL with TP53 disruption or del(11q), NOTCH1 mutations in +12 CLL conferred a significantly worse survival compared to that of +12 CLL with del(13q) or +12 only. The overrepresentation of cell cycle/proliferation related genes of +12 CLL with NOTCH1 mutations suggests the biological contribution of NOTCH1 mutations to determine a poor outcome. NOTCH1 mutations refine the intermediate prognosis of +12 CLL.


Blood | 2008

BCR ligation induced by IgM stimulation results in gene expression and functional changes only in IgVH unmutated chronic lymphocytic leukemia (CLL) cells

Anna Guarini; Sabina Chiaretti; Simona Tavolaro; Roberta Maggio; Nadia Peragine; Franca Citarella; Maria Rosaria Ricciardi; Simona Santangelo; Marilisa Marinelli; Maria Stefania De Propris; Monica Messina; Francesca Romana Mauro; Ilaria Del Giudice; R. Foa

Chronic lymphocytic leukemia (CLL) patients exhibit a variable clinical course. To investigate the association between clinicobiologic features and responsiveness of CLL cells to anti-IgM stimulation, we evaluated gene expression changes and modifications in cell-cycle distribution, proliferation, and apoptosis of IgV(H) mutated (M) and unmutated (UM) samples upon BCR cross-linking. Unsupervised analysis highlighted a different response profile to BCR stimulation between UM and M samples. Supervised analysis identified several genes modulated exclusively in the UM cases upon BCR cross-linking. Functional gene groups, including signal transduction, transcription, cell-cycle regulation, and cytoskeleton organization, were up-regulated upon stimulation in UM cases. Cell-cycle and proliferation analyses confirmed that IgM cross-linking induced a significant progression into the G(1) phase and a moderate increase of proliferative activity exclusively in UM patients. Moreover, we observed only a small reduction in the percentage of subG(0/1) cells, without changes in apoptosis, in UM cases; contrariwise, a significant increase of apoptotic levels was observed in stimulated cells from M cases. These results document that a differential genotypic and functional response to BCR ligation between IgV(H) M and UM cases is operational in CLL, indicating that response to antigenic stimulation plays a pivotal role in disease progression.


Haematologica | 2009

T-cell acute lymphoblastic leukemia

Sabina Chiaretti; Roberto Foa

The T-cell variant accounts for about 15 to 25% of acute lymphoblastic leukemias in children and adults. In this perspective article. Drs. Chiaretti and Foà examine our present understanding of this disorder. See related article on page 224.

Collaboration


Dive into the Sabina Chiaretti's collaboration.

Researchain Logo
Decentralizing Knowledge