Network


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

Hotspot


Dive into the research topics where Maria Zingariello is active.

Publication


Featured researches published by Maria Zingariello.


Haematologica | 2009

The JAK2V617 mutation induces constitutive activation and agonist hypersensitivity in basophils from patients with polycythemia vera

Lisa Pieri; Costanza Bogani; Paola Guglielmelli; Maria Zingariello; Rosa Alba Rana; Niccolò Bartalucci; Alberto Bosi; Alessandro M. Vannucchi

The JAK2 (V617F) mutation is found in almost all patients with polycythemia vera and an important fraction of patients with essential thrombocythemia and primary myelofibrosis. This study shows that basophil counts are increased in JAK2 (V617F)-positive patients, and that the basophils contain an increased number of granules. See related article on page 1484. Background The JAK2V617F mutation has been associated with constitutive and enhanced activation of neutrophils, while no information is available concerning other leukocyte subtypes. Design and Methods We evaluated correlations between JAK2V617F mutation and the count of circulating basophils, the number of activated CD63+ basophils, their response in vitro to agonists as well as the effects of a JAK2 inhibitor. Results We found that basophil count was increased in patients with JAK2V617F -positive myeloproliferative neoplasms, particularly in those with polycythemia vera, and was correlated with the V617F burden. The burden of V617F allele was similar in neutrophils and basophils from patients with polycythemia vera, while total JAK2 mRNA content was remarkably greater in the basophils; however, the content of JAK2 protein in basophils was not increased. The number of CD63+ basophils was higher in patients with polycythemia vera than in healthy subjects or patients with essential thrombocythemia or primary myelofibrosis and was correlated with the V617F burden. Ultrastructurally, basophils from patients with polycythemia vera contained an increased number of granules, most of which were empty suggesting cell degranulation in vivo. Ex vivo experiments revealed that basophils from patients with polycythemia vera were hypersensitive to the priming effect of interleukin-3 and to f-MLP-induced activation; pre-treatment with a JAK2 inhibitor reduced polycythemia vera basophil activation. Finally, we found that the number of circulating CD63+ basophils was significantly greater in patients suffering from aquagenic pruritus, who also showed a higher V617F allele burden. Conclusions These data indicate that the number of constitutively activated and hypersensitive circulating basophils is increased in polycythemia vera, underscoring a role of JAK2V617F in these cells’ abnormal function and, putatively, in the pathogenesis of pruritus.


Blood | 2013

Characterization of the TGF-β1 signaling abnormalities in the Gata1low mouse model of myelofibrosis.

Maria Zingariello; Fabrizio Martelli; Fiorella Ciaffoni; Francesca Masiello; Barbara Ghinassi; Emanuela D'Amore; Margherita Massa; Giovanni Barosi; Laura Sancillo; Xiaochun Li; Judith D. Goldberg; Rosa Alba Rana; Anna Rita Migliaccio

Primary myelofibrosis (PMF) is characterized by fibrosis, ineffective hematopoiesis in marrow, and hematopoiesis in extramedullary sites and is associated with abnormal megakaryocyte (MK) development and increased transforming growth factor (TGF)-β1 release. To clarify the role of TGF-β1 in the pathogenesis of this disease, the TGF-β1 signaling pathway of marrow and spleen of the Gata1(low) mouse model of myelofibrosis (MF) was profiled and the consequences of inhibition of TGF-β1 signaling on disease manifestations determined. The expression of 20 genes in marrow and 36 genes in spleen of Gata1(low) mice was altered. David-pathway analyses identified alterations of TGF-β1, Hedgehog, and p53 signaling in marrow and spleen and of mammalian target of rapamycin (mTOR) in spleen only and predicted that these alterations would induce consequences consistent with the Gata1(low) phenotype (increased apoptosis and G1 arrest both in marrow and spleen and increased osteoblast differentiation and reduced ubiquitin-mediated proteolysis in marrow only). Inhibition of TGF-β1 signaling normalized the expression of p53-related genes, restoring hematopoiesis and MK development and reducing fibrosis, neovascularization, and osteogenesis in marrow. It also normalized p53/mTOR/Hedgehog-related genes in spleen, reducing extramedullary hematopoiesis. These data identify altered expression signatures of TGF-β1 signaling that may be responsible for MF in Gata1(low) mice and may represent additional targets for therapeutic intervention in PMF.


Haematologica | 2015

Dexamethasone targeted directly to macrophages induces macrophage niches that promote erythroid expansion

Mario Falchi; Lilian Varricchio; Fabrizio Martelli; Francesca Masiello; Giulia Federici; Maria Zingariello; Gabriella Girelli; Carolyn Whitsett; Emanuel F. Petricoin; Søren K. Moestrup; Ann Zeuner; Anna Rita Migliaccio

Cultures of human CD34pos cells stimulated with erythroid growth factors plus dexamethasone, a model for stress erythropoiesis, generate numerous erythroid cells plus a few macrophages (approx. 3%; 3:1 positive and negative for CD169). Interactions occurring between erythroblasts and macrophages in these cultures and the biological effects associated with these interactions were documented by live phase-contrast videomicroscopy. Macrophages expressed high motility interacting with hundreds/thousands of erythroblasts per hour. CD169pos macrophages established multiple rapid ‘loose’ interactions with proerythroblasts leading to formation of transient erythroblastic island-like structures. By contrast, CD169neg macrophages established ‘tight’ interactions with mature erythroblasts and phagocytosed these cells. ‘Loose’ interactions of CD169pos macrophages were associated with proerythroblast cytokinesis (the M phase of the cell cycle) suggesting that these interactions may promote proerythroblast duplication. This hypothesis was tested by experiments that showed that as few as 103 macrophages significantly increased levels of 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide incorporation frequency in S/G2/M and cytokinesis expressed by proerythroblasts over 24 h of culture. These effects were observed also when macrophages were co-cultured with dexamethasone directly conjugated to a macrophage-specific CD163 antibody. In conclusion, in addition to promoting proerythroblast proliferation directly, dexamethasone stimulates expansion of these cells indirectly by stimulating maturation and cytokinesis supporting activity of macrophages.


Blood | 2009

Gata1 expression driven by the alternative HS2 enhancer in the spleen rescues the hematopoietic failure induced by the hypomorphic Gata1low mutation.

Anna Rita Migliaccio; Fabrizio Martelli; Maria Verrucci; Massimo Sanchez; Mauro Valeri; Giovanni Migliaccio; Alessandro M. Vannucchi; Maria Zingariello; Angela Di Baldassarre; Barbara Ghinassi; Rosa Alba Rana; Yvette van Hensbergen; Willem E. Fibbe

Rigorously defined reconstitution assays developed in recent years have allowed recognition of the delicate relationship that exists between hematopoietic stem cells and their niches. This balance ensures that hematopoiesis occurs in the marrow under steady-state conditions. However, during development, recovery from hematopoietic stress and in myeloproliferative disorders, hematopoiesis occurs in extramedullary sites whose microenvironments are still poorly defined. The hypomorphic Gata1(low) mutation deletes the regulatory sequences of the gene necessary for its expression in hematopoietic cells generated in the marrow. By analyzing the mechanism that rescues hematopoiesis in mice carrying this mutation, we provide evidence that extramedullary microenvironments sustain maturation of stem cells that would be otherwise incapable of maturing in the marrow.


Journal of Cellular Physiology | 2010

CXCR4-independent rescue of the myeloproliferative defect of the Gata1low myelofibrosis mouse model by Aplidin.

Maria Verrucci; Alessandro Pancrazzi; Miguel Aracil; Fabrizio Martelli; Paola Guglielmelli; Maria Zingariello; Barbara Ghinassi; Emanuela D'Amore; Jose Jimeno; Alessandro M. Vannucchi; Anna Rita Migliaccio

The discovery of JAK2 mutations in Philadelphia‐negative myeloproliferative neoplasms has prompted investigators to evaluate mutation‐targeted treatments to restore hematopoietic cell functions in these diseases. However, the results of the first clinical trials with JAK2 inhibitors are not as promising as expected, prompting a search for additional drugable targets to treat these disorders. In this paper, we used the hypomorphic Gata1low mouse model of primary myelofibrosis (PMF), the most severe of these neoplasms, to test the hypothesis that defective marrow hemopoiesis and development of extramedullary hematopoiesis in myelofibrosis is due to insufficient p27Kip1 activity and is treatable by Aplidin®, a cyclic depsipeptide that activates p27Kip1 in several cancer cells. Aplidin® restored expression of Gata1 and p27Kip1 in Gata1low hematopoietic cells, proliferation of marrow progenitor cells in vitro and maturation of megakaryocytes in vivo (reducing TGF‐β/VEGF levels released in the microenvironment by immature Gata1low megakaryocytes). Microvessel density, fibrosis, bone growth, and marrow cellularity were normal in Aplidin®‐treated mice and extramedullary hematopoiesis did not develop in liver although CXCR4 expression in Gata1low progenitor cells remained low. These results indicate that Aplidin® effectively alters the natural history of myelofibrosis in Gata1low mice and suggest this drug as candidate for clinical evaluation in PMF. J. Cell. Physiol. 225: 490–499, 2010.


RNA Biology | 2017

Absence of the Fragile X Mental Retardation Protein results in defects of RNA editing of neuronal mRNAs in mouse

Alice Filippini; Daniela Bonini; Caroline Lacoux; Laura Pacini; Maria Zingariello; Laura Sancillo; Daniela Bosisio; Valentina Salvi; Jessica Mingardi; Luca La Via; Francesca Zalfa; Claudia Bagni; Alessandro Barbon

ABSTRACT The fragile X syndrome (FXS), the most common form of inherited intellectual disability, is due to the absence of FMRP, a protein regulating RNA metabolism. Recently, an unexpected function of FMRP in modulating the activity of Adenosine Deaminase Acting on RNA (ADAR) enzymes has been reported both in Drosophila and Zebrafish. ADARs are RNA-binding proteins that increase transcriptional complexity through a post-transcriptional mechanism called RNA editing. To evaluate the ADAR2-FMRP interaction in mammals we analyzed several RNA editing re-coding sites in the fmr1 knockout (KO) mice. Ex vivo and in vitro analysis revealed that absence of FMRP leads to an increase in the editing levels of brain specific mRNAs, indicating that FMRP might act as an inhibitor of editing activity. Proximity Ligation Assay (PLA) in mouse primary cortical neurons and in non-neuronal cells revealed that ADAR2 and FMRP co-localize in the nucleus. The ADAR2-FMRP co-localization was further observed by double-immunogold Electron Microscopy (EM) in the hippocampus. Moreover, ADAR2-FMRP interaction appeared to be RNA independent. Because changes in the editing pattern are associated with neuropsychiatric and neurodevelopmental disorders, we propose that the increased editing observed in the fmr1-KO mice might contribute to the FXS molecular phenotypes.


Biotechnic & Histochemistry | 2009

Differential localization of P-selectin and von Willebrand factor during megakaryocyte maturation.

Maria Zingariello; Me Fabucci; Domenico Bosco; Anna Rita Migliaccio; Fabrizio Martelli; Ra Rana; E. Zetterberg

Abstract An important step in megakaryocyte maturation is the appropriate assembly of at least two distinct subsets of α-granules. The mechanism that sorts the α-granule components into distinct structures and mediates their release in response to specific stimuli is now emerging. P-selectin and von Willebrand factor are two proteins present in the α-granules that recognize P-selectin glycoprotein ligand on neutrophils and collagen in the subendothelial matrix. These proteins may play an important role in determining the differential release of the α-granule contents in response to external stimuli. If P-selectin and von Willebrand factor are localized in the same or different α-granules is not known. To clarify this question, we analyzed by immunoelectron microscopy the localization of von Willebrand factor and P-selectin during the maturation of wild-type and Gata1low megakaryocytes induced in vivo by treating animals with thrombopoietin. Gata1low is a hypomorphic mutation that blocks megakaryocyte maturation, reduces the levels of von Willebrand factor expression and displaces P-selectin on the demarcation membrane system. The maturation block induced by this mutation is partially rescued by treatment in vivo with thrombopoietin. In immature megakaryocytes, both wild-type and Gata1low, the two receptors were co-localized in the same cytoplasmic structures. By contrast, the two proteins were segregated to separate α-granule subsets as the megakaryocytes matured. These observations support the hypothesis that P-selectin and von Willebrand factor may ensure differential release of the α-granule content in response to external stimuli.


Annals of the New York Academy of Sciences | 2009

Removal of the spleen in mice alters the cytokine expression profile of the marrow micro-environment and increases bone formation.

Fabrizio Martelli; Maria Verrucci; Giovanni Migliaccio; Maria Zingariello; Rosa Alba Rana; Alessandro M. Vannucchi; Anna Rita Migliaccio

Splenectomized mice express progressively increased numbers of platelets in the blood and reduced numbers of megakaryocytes in the marrow with age. The megakaryocytes in the marrow of these animals express reduced levels of Gata1, a transcription factor necessary for their maturation. In addition, the marrow from these animals expresses greater levels of cytokines (TGF‐β, PDGF‐α, and VEGF) known to be produced at high levels by megakaryocytes expressing reduced levels of Gata1. This high level of cytokine expression is in turn associated with active osteoblast proliferation localized to areas of the femur, where megakaryocytes expressing reduced Gata1 levels are also found. These results confirm the role of megakaryocytes as regulator of bone formation in mice and suggest that a cross‐talk between the spleen and marrow may regulate the total numbers of hemopoietic niches present in an animal.


Molecular Carcinogenesis | 2017

IGFBP-3 inhibits Wnt signaling in metastatic melanoma cells

Antimo Naspi; Maria Zingariello; Laura Sancillo; Vincenzo Panasiti; Dorina Polinari; Marianna Martella; Rana Rosa Alba; Paola Londei

In previous works, we have shown that insulin‐like growth factor‐binding protein‐3 (IGFBP‐3), a tissue and circulating protein able to bind to IGFs, decreases drastically in the blood serum of patients with diffuse metastatic melanoma. In agreement with the clinical data, recombinant IGFBP‐3 was found to inhibit the motility and invasiveness of cultured metastatic melanoma cells and to prevent growth of grafted melanomas in mice. The present work was aimed at identifying the signal transduction pathways underlying the anti‐tumoral effects of IGFBP‐3. We show that the anti‐tumoral effect of IGFBP‐3 is due to inhibition of the Wnt pathway and depends upon the presence of CD44, a receptor protein known to modulate Wnt signaling. Once it has entered the cell, IGFBP‐3 binds the Wnt signalosome interacting specifically with its component GSK‐3β. As a consequence, the β‐catenin destruction complex dissociates from the LRP6 Wnt receptor and GSK‐3β is activated through dephosphorylation, becoming free to target cytoplasmic β‐catenin which is degraded by the proteasomal pathway. Altogether, the results suggest that IGFBP‐3 is a novel and effective inhibitor of Wnt signaling. As IGFBP‐3 is a physiological protein which has no detectable toxic effects either on cultured cells or live mice, it might qualify as an interesting new therapeutic agent in melanoma, and potentially many other cancers with a hyperactive Wnt signaling.


Stem Cells | 2016

P‐Selectin Sustains Extramedullary Hematopoiesis in the Gata1low Model of Myelofibrosis

Gerald J. Spangrude; Daniel Lewandowski; Fabrizio Martelli; Manuela Marra; Maria Zingariello; Laura Sancillo; Rosa Alba Rana; Anna Rita Migliaccio

Splenomegaly is a major manifestation of primary myelofibrosis (PMF) contributing to clinical symptoms and hematologic abnormalities. The spleen from PMF patients contains increased numbers of hematopoietic stem cells (HSC) and megakaryocytes (MK). These MK express high levels of P‐selectin (P‐sel) that, by triggering neutrophil emperipolesis, may cause TGF‐β release and disease progression. This hypothesis was tested by deleting the P‐sel gene in the myelofibrosis mouse model carrying the hypomorphic Gata1low mutation that induces megakaryocyte abnormalities that recapitulate those observed in PMF. P‐selnullGata1low mice survived splenectomy and lived 3 months longer than P‐selWTGata1low littermates and expressed limited fibrosis and osteosclerosis in the marrow or splenomegaly. Furthermore, deletion of P‐sel disrupted megakaryocyte/neutrophil interactions in spleen, reduced TGF‐β content, and corrected the HSC distribution that in Gata1low mice, as in PMF patients, is abnormally expanded in spleen. Conversely, pharmacological inhibition of TGF‐β reduced P‐sel expression in MK and corrected HSC distribution. Spleens, but not marrow, of Gata1low mice contained numerous cKITpos activated fibrocytes, probably of dendritic cell origin, whose membrane protrusions interacted with MK establishing niches hosting immature cKITpos hematopoietic cells. These activated fibrocytes were not detected in spleens from P‐selnullGata1low or TGF‐β‐inhibited Gata1low littermates and were observed in spleen, but not in marrow, from PMF patients. Therefore, in Gata1low mice, and possibly in PMF, abnormal P‐sel expression in MK may mediate the pathological cell interactions that increase TGF‐β content in MK and favor establishment of a microenvironment that supports myelofibrosis‐related HSC in spleen. Stem Cells 2016;34:67–82

Collaboration


Dive into the Maria Zingariello's collaboration.

Top Co-Authors

Avatar

Anna Rita Migliaccio

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Rosa Alba Rana

University of Chieti-Pescara

View shared research outputs
Top Co-Authors

Avatar

Fabrizio Martelli

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Laura Sancillo

University of Chieti-Pescara

View shared research outputs
Top Co-Authors

Avatar

Barbara Ghinassi

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Verrucci

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emanuela D'Amore

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Francesca Masiello

Istituto Superiore di Sanità

View shared research outputs
Researchain Logo
Decentralizing Knowledge