Network


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

Hotspot


Dive into the research topics where Valentina Pennucci is active.

Publication


Featured researches published by Valentina Pennucci.


Blood | 2014

miRNA-mRNA integrative analysis in primary myelofibrosis CD34+ cells: role of miR-155/JARID2 axis in abnormal megakaryopoiesis

Ruggiero Norfo; Roberta Zini; Valentina Pennucci; Elisa Bianchi; Simona Salati; Paola Guglielmelli; Costanza Bogani; Tiziana Fanelli; Carmela Mannarelli; Vittorio Rosti; Daniela Pietra; Silvia Salmoiraghi; Andrea Bisognin; Samantha Ruberti; Sebastiano Rontauroli; Giorgia Sacchi; Zelia Prudente; Giovanni Barosi; Mario Cazzola; Alessandro Rambaldi; Stefania Bortoluzzi; Sergio Ferrari; Enrico Tagliafico; Alessandro M. Vannucchi; Rossella Manfredini

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by megakaryocyte (MK) hyperplasia, bone marrow fibrosis, and abnormal stem cell trafficking. PMF may be associated with somatic mutations in JAK2, MPL, or CALR. Previous studies have shown that abnormal MKs play a central role in the pathophysiology of PMF. In this work, we studied both gene and microRNA (miRNA) expression profiles in CD34(+) cells from PMF patients. We identified several biomarkers and putative molecular targets such as FGR, LCN2, and OLFM4. By means of miRNA-gene expression integrative analysis, we found different regulatory networks involved in the dysregulation of transcriptional control and chromatin remodeling. In particular, we identified a network gathering several miRNAs with oncogenic potential (eg, miR-155-5p) and targeted genes whose abnormal function has been previously associated with myeloid neoplasms, including JARID2, NR4A3, CDC42, and HMGB3. Because the validation of miRNA-target interactions unveiled JARID2/miR-155-5p as the strongest relationship in the network, we studied the function of this axis in normal and PMF CD34(+) cells. We showed that JARID2 downregulation mediated by miR-155-5p overexpression leads to increased in vitro formation of CD41(+) MK precursors. These findings suggest that overexpression of miR-155-5p and the resulting downregulation of JARID2 may contribute to MK hyperplasia in PMF.


Cell Death & Differentiation | 2015

MYB controls erythroid versus megakaryocyte lineage fate decision through the miR-486-3p-mediated downregulation of MAF.

Elisa Bianchi; Jenny Bulgarelli; Samantha Ruberti; Sebastiano Rontauroli; Giorgia Sacchi; Ruggiero Norfo; Valentina Pennucci; Roberta Zini; Simona Salati; Zelia Prudente; Sergio Ferrari; Rossella Manfredini

The transcription factor MYB has a key role in hematopoietic progenitor cells (HPCs) lineage choice, by enhancing erythropoiesis at the expense of megakaryopoiesis. We previously demonstrated that MYB controls erythroid versus megakaryocyte lineage decision by transactivating KLF1 and LMO2 expression. To further unravel the molecular mechanisms through which MYB affects lineage fate decision, we performed the integrative analysis of miRNA and mRNA changes in MYB-silenced human primary CD34+ HPCs. Among the miRNAs with the highest number of predicted targets, we focused our studies on hsa-miR-486-3p by demonstrating that MYB controls miR-486-3p expression through the transactivation of its host gene, ankyrin-1 (ANK1) and that miR-486-3p affects HPCs commitment. Indeed, overexpression and knockdown experiments demonstrated that miR-486-3p supports the erythropoiesis while restraining the megakaryopoiesis. Of note, miR-486-3p also favors granulocyte differentiation while repressing the macrophage differentiation. To shed some light on the molecular mechanisms through which miR-486-3p affects HPCs lineage commitment, we profiled the gene expression changes upon miR-486-3p overexpression in CD34+ cells. Among the genes downregulated in miR-486-3p-overexpressing HPCs and computationally predicted to be miR-486-3p targets, we identified MAF as a miR-486-3p target by 3′UTR luciferase reporter assay. Noteworthy, MAF overexpression was able to partially reverse the effects of miR-486-3p overexpression on erythroid versus megakaryocyte lineage choice. Moreover, the MYB/MAF co-silencing constrained the skewing of erythroid versus megakaryocyte lineage commitment in MYB-silenced CD34+ cells, by restraining the expansion of megakaryocyte lineage while partially rescuing the impairment of erythropoiesis. Therefore, our data collectively demonstrate that MYB favors erythropoiesis and restrains megakaryopoiesis through the transactivation of miR-486-3p expression and the subsequent downregulation of MAF. As a whole, our study uncovers the MYB/miR-486-3p/MAF axis as a new mechanism underlying the MYB-driven control of erythroid versus megakaryocyte lineage fate decision.


Blood | 2016

Genomic landscape of megakaryopoiesis and platelet function defects

Elisa Bianchi; Ruggiero Norfo; Valentina Pennucci; Roberta Zini; Rossella Manfredini

Megakaryopoiesis is a complex, stepwise process that takes place largely in the bone marrow. At the apex of the hierarchy, hematopoietic stem cells undergo a number of lineage commitment decisions that ultimately lead to the production of polyploid megakaryocytes. On average, megakaryocytes release 10(11) platelets per day into the blood that repair vascular injuries and prevent excessive bleeding. This differentiation process is tightly controlled by exogenous and endogenous factors, which have been the topics of intense research in the hematopoietic field. Indeed, a skewing of megakaryocyte commitment and differentiation may entail the onset of myeloproliferative neoplasms and other preleukemic disorders together with acute megakaryoblastic leukemia, whereas quantitative or qualitative defects in platelet production can lead to inherited platelet disorders. The recent advent of next-generation sequencing has prompted mapping of the genomic landscape of these conditions to provide an accurate view of the underlying lesions. The aims of this review are to introduce the physiological pathways of megakaryopoiesis and to present landmark studies on acquired and inherited disorders that target them. These studies have not only introduced a new era in the fields of molecular medicine and targeted therapies but may also provide us with a better understanding of the mechanisms underlying normal megakaryopoiesis and thrombopoiesis that can inform efforts to create alternative sources of megakaryocytes and platelets.


PLOS ONE | 2013

Co-Culture of Hematopoietic Stem/Progenitor Cells with Human Osteblasts Favours Mono/Macrophage Differentiation at the Expense of the Erythroid Lineage

Simona Salati; Gina Lisignoli; Cristina Manferdini; Valentina Pennucci; Roberta Zini; Elisa Bianchi; Ruggiero Norfo; Andrea Facchini; Sergio Ferrari; Rossella Manfredini

Hematopoietic stem cells (HSCs) are located in the bone marrow in a specific microenvironment referred as the hematopoietic stem cell niche, where HSCs interact with a variety of stromal cells. Though several components of the stem cell niche have been identified, the regulatory mechanisms through which such components regulate the stem cell fate are still unknown. In order to address this issue, we investigated how osteoblasts (OBs) can affect the molecular and functional phenotype of Hematopoietic Stem/Progenitor Cells (HSPCs) and vice versa. For this purpose, human CD34+ cells were cultured in direct contact with primary human OBs. Our data showed that CD34+ cells cultured with OBs give rise to higher total cell numbers, produce more CFUs and maintain a higher percentage of CD34+CD38- cells compared to control culture. Moreover, clonogenic assay and long-term culture results showed that co-culture with OBs induces a strong increase in mono/macrophage precursors coupled to a decrease in the erythroid ones. Finally, gene expression profiling (GEP) allowed us to study which signalling pathways were activated in the hematopoietic cell fraction and in the stromal cell compartment after coculture. Such analysis allowed us to identify several cytokine-receptor networks, such as WNT pathway, and transcription factors, as TWIST1 and FOXC1, that could be activated by co-culture with OBs and could be responsible for the biological effects reported above. Altogether our results indicate that OBs are able to affect HPSCs on 2 different levels: on one side, they increase the immature progenitor pool in vitro, on the other side, they favor the expansion of the mono/macrophage precursors at the expense of the erythroid lineage.


Experimental Hematology | 2012

Valproic acid triggers erythro/megakaryocyte lineage decision through induction of GFI1B and MLLT3 expression.

Roberta Zini; Ruggiero Norfo; Francesco Ferrari; Elisa Bianchi; Simona Salati; Valentina Pennucci; Giorgia Sacchi; Chiara Carboni; Giovanni Battista Ceccherelli; Enrico Tagliafico; Sergio Ferrari; Rossella Manfredini

Histone deacetylase inhibitors represent a family of targeted anticancer compounds that are widely used against hematological malignancies. So far little is known about their effects on normal myelopoiesis. Therefore, in order to investigate the effect of histone deacetylase inhibitors on the myeloid commitment of hematopoietic stem/progenitor cells, we treated CD34(+) cells with valproic acid (VPA). Our results demonstrate that VPA treatment induces H4 histone acetylation and hampers cell cycle progression in CD34(+) cells sustaining high levels of CD34 protein expression. In addition, our data show that VPA treatment promotes erythrocyte and megakaryocyte differentiation. In fact, we demonstrate that VPA treatment is able to induce the expression of growth factor-independent protein 1B (GFI1B) and of mixed-lineage leukemia translocated to chromosome 3 protein (MLLT3), which are crucial regulators of erythrocyte and megakaryocyte differentiation, and that the up-regulation of these genes is mediated by the histone hyperacetylation at their promoter sites. Finally, we show that GFI1B inhibition impairs erythroid and megakaryocyte differentiation induced by VPA, while MLLT3 silencing inhibits megakaryocyte commitment only. As a whole, our data suggest that VPA sustains the expression of stemness-related markers in hematopoietic stem/progenitor cells and is able to interfere with hematopoietic lineage commitment by enhancing erythrocyte and megakaryocyte differentiation and by inhibiting the granulocyte and mono-macrophage maturation.


Leukemia & Lymphoma | 2015

Abnormal expression patterns of WT1-as, MEG3 and ANRIL long non-coding RNAs in CD34+ cells from patients with primary myelofibrosis and their clinical correlations

Valentina Pennucci; Roberta Zini; Ruggiero Norfo; Paola Guglielmelli; Elisa Bianchi; Simona Salati; Giorgia Sacchi; Zelia Prudente; Elena Tenedini; Samantha Ruberti; Chiara Paoli; Tiziana Fanelli; Carmela Mannarelli; Enrico Tagliafico; Sergio Ferrari; Alessandro M. Vannucchi; Rossella Manfredini

Valentina Pennucci 1 , Roberta Zini 1 , Ruggiero Norfo 1 , Paola Guglielmelli 2 , Elisa Bianchi 1 , Simona Salati 1 , Giorgia Sacchi 1 , Zelia Prudente 1 , Elena Tenedini 3 , Samantha Ruberti 1 , Chiara Paoli 2 , Tiziana Fanelli 2 , Carmela Mannarelli 2 , Enrico Tagliafi co 3 , Sergio Ferrari 3 , Alessandro M. Vannucchi 2 , Rossella Manfredini 1 & on behalf of Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative (AGIMM) Investigators


Blood Cancer Journal | 2016

A data-driven network model of primary myelofibrosis: transcriptional and post-transcriptional alterations in CD34+ cells

Enrica Calura; Silvia Pizzini; Andrea Bisognin; Alessandro Coppe; Gabriele Sales; Enrico Gaffo; Tiziana Fanelli; Carmela Mannarelli; Roberta Zini; Ruggiero Norfo; Valentina Pennucci; Rossella Manfredini; Chiara Romualdi; Paola Guglielmelli; Alessandro M. Vannucchi; Stefania Bortoluzzi

microRNAs (miRNAs) are relevant in the pathogenesis of primary myelofibrosis (PMF) but our understanding is limited to specific target genes and the overall systemic scenario islacking. By both knowledge-based and ab initio approaches for comparative analysis of CD34+ cells of PMF patients and healthy controls, we identified the deregulated pathways involving miRNAs and genes and new transcriptional and post-transcriptional regulatory circuits in PMF cells. These converge in a unique and integrated cellular process, in which the role of specific miRNAs is to wire, co-regulate and allow a fine crosstalk between the involved processes. The PMF pathway includes Akt signaling, linked to Rho GTPases, CDC42, PLD2, PTEN crosstalk with the hypoxia response and Calcium-linked cellular processes connected to cyclic AMP signaling. Nested on the depicted transcriptional scenario, predicted circuits are reported, opening new hypotheses. Links between miRNAs (miR-106a-5p, miR-20b-5p, miR-20a-5p, miR-17-5p, miR-19b-3p and let-7d-5p) and key transcription factors (MYCN, ATF, CEBPA, REL, IRF and FOXJ2) and their common target genes tantalizingly suggest new path to approach the disease. The study provides a global overview of transcriptional and post-transcriptional deregulations in PMF, and, unifying consolidated and predicted data, could be helpful to identify new combinatorial therapeutic strategy. Interactive PMF network model: http://compgen.bio.unipd.it/pmf-net/.


Leukemia | 2018

Involvement of MAF/SPP1 axis in the development of bone marrow fibrosis in PMF patients

Samantha Ruberti; Elisa Bianchi; Paola Guglielmelli; Sebastiano Rontauroli; Greta Barbieri; L Tavernari; Tiziana Fanelli; Ruggiero Norfo; Valentina Pennucci; Giuditta Corbizi Fattori; Carmela Mannarelli; Niccolò Bartalucci; Barbara Mora; L Elli; Maria Antonietta Avanzini; Chiara Rossi; Silvia Salmoiraghi; Roberta Zini; Simona Salati; Zelia Prudente; Vittorio Rosti; Francesco Passamonti; Alessandro Rambaldi; Sergio Ferrari; Enrico Tagliafico; Alessandro M. Vannucchi; Rossella Manfredini

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by hyperplastic megakaryopoiesis and myelofibrosis. We recently described the upregulation of MAF (v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog) in PMF CD34+ hematopoietic progenitor cells (HPCs) compared to healthy donor. Here we demonstrated that MAF is also upregulated in PMF compared with the essential thrombocytemia (ET) and polycytemia vera (PV) HPCs. MAF overexpression and knockdown experiments shed some light into the role of MAF in PMF pathogenesis, by demonstrating that MAF favors the megakaryocyte and monocyte/macrophage commitment of HPCs and leads to the increased expression of proinflammatory and profibrotic mediators. Among them, we focused our further studies on SPP1 and LGALS3. We assessed SPP1 and LGALS3 protein levels in 115 PMF, 47 ET and 24 PV patients plasma samples and we found that SPP1 plasma levels are significantly higher in PMF compared with ET and PV patients. Furthermore, in vitro assays demonstrated that SPP1 promotes fibroblasts and mesenchymal stromal cells proliferation and collagen production. Strikingly, clinical correlation analyses uncovered that higher SPP1 plasma levels in PMF patients correlate with a more severe fibrosis degree and a shorter overall survival. Collectively our data unveil that MAF overexpression contributes to PMF pathogenesis by driving the deranged production of the profibrotic mediator SPP1.


Oncotarget | 2017

miR-494-3p overexpression promotes megakaryocytopoiesis in primary myelofibrosis hematopoietic stem/progenitor cells by targeting SOCS6

Sebastiano Rontauroli; Ruggiero Norfo; Valentina Pennucci; Roberta Zini; Samantha Ruberti; Elisa Bianchi; Simona Salati; Zelia Prudente; Chiara Rossi; Vittorio Rosti; Paola Guglielmelli; Giovanni Barosi; Alessandro M. Vannucchi; Enrico Tagliafico; Rossella Manfredini

Primary myelofibrosis (PMF) is a chronic Philadelphia-negative myeloproliferative neoplasm characterized by hematopoietic stem cell-derived clonal myeloproliferation, involving especially the megakaryocyte lineage. To better characterize how the altered expression of microRNAs might contribute to PMF pathogenesis, we have previously performed the integrative analysis of gene and microRNA expression profiles of PMF hematopoietic stem/progenitor cells (HSPCs), which allowed us to identify miR-494-3p as the upregulated microRNA predicted to target the highest number of downregulated mRNAs. To elucidate the role of miR-494-3p in hematopoietic differentiation, in the present study we demonstrated that miR-494-3p enforced expression in normal HSPCs promotes megakaryocytopoiesis. Gene expression profiling upon miR-494-3p overexpression allowed the identification of genes commonly downregulated both after microRNA overexpression and in PMF CD34+ cells. Among them, suppressor of cytokine signaling 6 (SOCS6) was confirmed to be a miR-494-3p target by luciferase assay. Western blot analysis showed reduced level of SOCS6 protein as well as STAT3 activation in miR-494-3p overexpressing cells. Furthermore, transient inhibition of SOCS6 expression in HSPCs demonstrated that SOCS6 silencing stimulates megakaryocytopoiesis, mimicking the phenotypic effects observed upon miR-494-3p overexpression. Finally, to disclose the contribution of miR-494-3p upregulation to PMF pathogenesis, we performed inhibition experiments in PMF HSPCs, which showed that miR-494-3p silencing led to SOCS6 upregulation and impaired megakaryocyte differentiation. Taken together, our results describe for the first time the role of miR-494-3p during normal HSPC differentiation and suggest that its increased expression, and the subsequent downregulation of its target SOCS6, might contribute to the megakaryocyte hyperplasia commonly observed in PMF patients.


International Journal of Cancer | 2016

Integrative analysis of copy number and gene expression data suggests novel pathogenetic mechanisms in primary myelofibrosis

Simona Salati; Roberta Zini; Simona Nuzzo; Paola Guglielmelli; Valentina Pennucci; Zelia Prudente; Samantha Ruberti; Sebastiano Rontauroli; Ruggiero Norfo; Elisa Bianchi; Costanza Bogani; Giada Rotunno; Tiziana Fanelli; Carmela Mannarelli; Vittorio Rosti; Silvia Salmoiraghi; Daniela Pietra; Sergio Ferrari; Giovanni Barosi; Alessandro Rambaldi; Mario Cazzola; Silvio Bicciato; Enrico Tagliafico; Alessandro M. Vannucchi; Rossella Manfredini

Collaboration


Dive into the Valentina Pennucci's collaboration.

Top Co-Authors

Avatar

Rossella Manfredini

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Ruggiero Norfo

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Elisa Bianchi

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Roberta Zini

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Simona Salati

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samantha Ruberti

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Sergio Ferrari

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Enrico Tagliafico

University of Modena and Reggio Emilia

View shared research outputs
Researchain Logo
Decentralizing Knowledge