Network


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

Hotspot


Dive into the research topics where Saula Checquolo is active.

Publication


Featured researches published by Saula Checquolo.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Combined expression of pTα and Notch3 in T cell leukemia identifies the requirement of preTCR for leukemogenesis

Diana Bellavia; Antonio Francesco Campese; Saula Checquolo; Anna Balestri; Andrea Biondi; Giovanni Cazzaniga; Urban Lendahl; Hans Jörg Fehling; Adrian Hayday; Luigi Frati; Harald von Boehmer; Alberto Gulino; Isabella Screpanti

Notch receptors are conserved regulators of cell fate and have been implicated in the regulation of T cell differentiation and lymphomagenesis. However, neither the generality of Notch involvement in leukemia, nor the molecules with which Notch may interact have been clarified. Recently, we showed that transgenic mice expressing the constitutively active intracellular domain of Notch3 in thymocytes and T cells developed early and aggressive T cell neoplasias. Although primarily splenic, the tumors sustained features of immature thymocytes, including expression of pTα, a defining component of the pre T cell receptor, known to be a potent signaling complex provoking thymocyte survival, proliferation, and activation. Thus, enforced expression of Notch3, which is ordinarily down-regulated as thymocytes mature, may sustain pre T cell receptor expression, causing dysregulated hyperplasia. This hypothesis has been successfully tested in this article by the observation that deletion of pTα in Notch3 transgenic mice abrogates tumor development, indicating a crucial role for pTα in T cell leukemogenesis. Parallel observations were made in humans, in that all T cell acute lymphoblastic leukemias examined showed expression of Notch3 and of the Notch target gene HES-1, as well as of pTα a and b transcripts, whereas the expression of all these genes was dramatically reduced or absent in remission. Together, these results suggest that the combined expression of Notch3 and pTα sustains T cell leukemogenesis and may represent pathognomonic molecular features of human T-ALL.


American Journal of Human Genetics | 2010

Heterozygous Germline Mutations in the CBL Tumor-Suppressor Gene Cause a Noonan Syndrome-like Phenotype

Simone Martinelli; Alessandro De Luca; Emilia Stellacci; Cesare Rossi; Saula Checquolo; Francesca Lepri; Viviana Caputo; Marianna Silvano; Francesco Buscherini; Federica Consoli; Grazia Ferrara; Maria Cristina Digilio; Maria Luigia Cavaliere; Johanna M. van Hagen; Giuseppe Zampino; Ineke van der Burgt; Giovanni Battista Ferrero; Laura Mazzanti; Isabella Screpanti; Helger G. Yntema; Willy M. Nillesen; Ravi Savarirayan; Martin Zenker; Bruno Dallapiccola; Bruce D. Gelb; Marco Tartaglia

RAS signaling plays a key role in controlling appropriate cell responses to extracellular stimuli and participates in early and late developmental processes. Although enhanced flow through this pathway has been established as a major contributor to oncogenesis, recent discoveries have revealed that aberrant RAS activation causes a group of clinically related developmental disorders characterized by facial dysmorphism, a wide spectrum of cardiac disease, reduced growth, variable cognitive deficits, ectodermal and musculoskeletal anomalies, and increased risk for certain malignancies. Here, we report that heterozygous germline mutations in CBL, a tumor-suppressor gene that is mutated in myeloid malignancies and encodes a multivalent adaptor protein with E3 ubiquitin ligase activity, can underlie a phenotype with clinical features fitting or partially overlapping Noonan syndrome (NS), the most common condition of this disease family. Independent CBL mutations were identified in two sporadic cases and two families from among 365 unrelated subjects who had NS or suggestive features and were negative for mutations in previously identified disease genes. Phenotypic heterogeneity and variable expressivity were documented. Mutations were missense changes altering evolutionarily conserved residues located in the RING finger domain or the linker connecting this domain to the N-terminal tyrosine kinase binding domain, a known mutational hot spot in myeloid malignancies. Mutations were shown to affect CBL-mediated receptor ubiquitylation and dysregulate signal flow through RAS. These findings document that germline mutations in CBL alter development to cause a clinically variable condition that resembles NS and that possibly predisposes to malignancies.


Journal of Immunology | 2003

Expression of Activated Notch3 in Transgenic Mice Enhances Generation of T Regulatory Cells and Protects against Experimental Autoimmune Diabetes

Emanuela Anastasi; Antonio Francesco Campese; Diana Bellavia; Angela Bulotta; Anna Balestri; Monica Pascucci; Saula Checquolo; Roberto Gradini; Urban Lendahl; Luigi Frati; Alberto Gulino; Umberto Di Mario; Isabella Screpanti

Thymic-derived dysregulated tolerance has been suggested to occur in type 1 diabetes via impaired generation of CD4+CD25+ T regulatory cells, leading to autoimmune β cell destruction. In this study, we demonstrate that Notch3 expression is a characteristic feature of CD4+CD25+ cells. Furthermore, streptozotocin-induced autoimmune diabetes fails to develop in transgenic mice carrying the constitutively active intracellular domain of Notch3 in thymocytes and T cells. The failure to develop the disease is associated with an increase of CD4+CD25+ T regulatory cells, accumulating in lymphoid organs, in pancreas infiltrates and paralleled by increased expression of IL-4 and IL-10. Accordingly, CD4+ T cells from Notch3-transgenic mice inhibit the development of hyperglycemia and insulitis when injected into streptozotocin-treated wild-type mice and display in vitro suppressive activity. These observations, therefore, suggest that Notch3-mediated events regulate the expansion and function of T regulatory cells, leading to protection from experimental autoimmune diabetes and identify the Notch pathway as a potential target for therapeutic intervention in type 1 diabetes.


Leukemia | 2014

Notch and NF-kB signaling pathways regulate miR-223/FBXW7 axis in T-cell acute lymphoblastic leukemia.

V. Kumar; Rocco Palermo; Claudio Talora; Antonio Francesco Campese; Saula Checquolo; Diana Bellavia; L. Tottone; G. Testa; Ermanno Miele; S. Indraccolo; A. Amadori; Elisabetta Ferretti; Alberto Gulino; Alessandra Vacca; Isabella Screpanti

Notch signaling deregulation is linked to the onset of several tumors including T-cell acute lymphoblastic leukemia (T-ALL). Deregulated microRNA (miRNA) expression is also associated with several cancers, including leukemias. However, the transcriptional regulators of miRNAs, as well as the relationships between Notch signaling and miRNA deregulation, are poorly understood. To identify miRNAs regulated by Notch pathway, we performed microarray-based miRNA profiling of several Notch-expressing T-ALL models. Among seven miRNAs, consistently regulated by overexpressing or silencing Notch3, we focused our attention on miR-223, whose putative promoter analysis revealed a conserved RBPjk binding site, which was nested to an NF-kB consensus. Luciferase and chromatin immunoprecipitation assays on the promoter region of miR-223 show that both Notch and NF-kB are novel coregulatory signals of miR-223 expression, being able to activate cooperatively the transcriptional activity of miR-223 promoter. Notably, the Notch-mediated activation of miR-223 represses the tumor suppressor FBXW7 in T-ALL cell lines. Moreover, we observed the inverse correlation of miR-223 and FBXW7 expression in a panel of T-ALL patient-derived xenografts. Finally, we show that miR-223 inhibition prevents T-ALL resistance to γ-secretase inhibitor (GSI) treatment, suggesting that miR-223 could be involved in GSI sensitivity and its inhibition may be exploited in target therapy protocols.


EMBO Reports | 2003

Pre‐TCR‐triggered ERK signalling‐dependent downregulation of E2A activity in Notch3‐induced T‐cell lymphoma

Claudio Talora; Antonio Francesco Campese; Diana Bellavia; Monica Pascucci; Saula Checquolo; Manuela Groppioni; Luigi Frati; Harald von Boehmer; Alberto Gulino; Isabella Screpanti

Notch and basic helix–loop–helix E2A pathways specify cell fate and regulate neoplastic transformation in a variety of cell types. Whereas Notch enhances tumorigenesis, E2A suppresses it. However, whether and how Notch and E2A interact functionally in an integrative mechanism for regulating neoplastic transformation remains to be understood. It has been shown that Notch3‐induced T‐cell leukaemia is abrogated by the inactivation of pTα/pre‐T‐cell antigen receptor (pre‐TCR). We report here that Notch3‐induced transcriptional activation of pTα/pre‐TCR is responsible for the downregulation of E2A DNA binding and transcriptional activity. Further, the E2A messenger RNA and protein levels remain unaltered but the E2A inhibitor Id1 expression is augmented in thymocytes and T lymphoma cells derived from Notch3 transgenic mice. The increase in Id1 expression is achieved by pre‐TCR‐induced extracellular‐signalling‐regulated kinase 1/2. These observations support a model in which the upregulation of pre‐TCR signalling seems to be the prerequi‐site for Notch3‐induced inhibition of E2A, thus leading to the development of lymphoma in Notch3 transgenic mice.


Oncogene | 2008

Notch3: from subtle structural differences to functional diversity.

Diana Bellavia; Saula Checquolo; Antonio Francesco Campese; Maria Pia Felli; Alberto Gulino; Isabella Screpanti

The Notch3 gene was identified, at the beginning of 90s, as the third mammalian Notch and was initially reported as being expressed in proliferating neuroepithelium. Since then, increasing evidence has demonstrated a number of structural and functional differences between Notch3 and both Notch1 and Notch2, which exhibit the highest structural similarity among the four mammalian Notch receptors. Possibly due to its more restricted tissue distribution, targeted deletion of murine Notch3 does not lead to embryonic lethality as is observed with targeted deletion of Notch1 and Notch2. However, genetic mutation, amplification and deregulated expression of Notch3 have been correlated with the disruption of cell differentiation in transgenic mice and to development of diseases in mice and humans. This review discusses the possible relationships between the structural differences and the nonredundant roles that Notch3 plays in the pathogenesis of the human disease cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy and in the regulation of murine T-cell differentiation and leukemogenesis.


Oncogene | 2005

PKC theta mediates pre-TCR signaling and contributes to Notch3-induced T-cell leukemia.

Maria Pia Felli; Alessandra Vacca; Angelica Calce; Diana Bellavia; Antonio Francesco Campese; Rita Luciana Grillo; Monica Di Giovine; Saula Checquolo; Claudio Talora; Rocco Palermo; Giuseppina Di Mario; Luigi Frati; Alberto Gulino; Isabella Screpanti

Protein kinase (PK)Cθ is a critical regulator of mature T-cell activation and proliferation, being implicated in TCR-triggered nuclear factor (NF)-κB activation and providing important survival signals to leukemic T cells. We previously showed that overexpression of pTα/pre-TCR and constitutive activation of NF-κB characterize the T-cell leukemia/lymphoma developing in Notch3-IC transgenic mice. We report here that PKCθ is a downstream target of Notch3 signaling and that its activation and membrane translocation require a functional pre-TCR in order to trigger NF-κB activation in thymocytes and lymphoma cells of transgenic mice. Furthermore, deletion of PKCθ in Notch3-IC transgenic mice reduces the incidence of leukemia, correlating with decreased NF-κB activation. This paper therefore suggests that PKCθ mediates the activation of NF-κB by pre-TCR in immature thymocytes and contributes to the development of Notch3-dependent T-cell lymphoma.


Oncogene | 2012

Acetylation controls Notch3 stability and function in T-cell leukemia

Rocco Palermo; Saula Checquolo; A Giovenco; Paola Grazioli; V Kumar; Antonio Francesco Campese; A Giorgi; Maddalena Napolitano; Gianluca Canettieri; Grazia Ferrara; M E Schininà; Marella Maroder; Luigi Frati; Alberto Gulino; Alessandra Vacca; Isabella Screpanti

Post-translational modifications of Notch3 and their functional role with respect to Notch3 overexpression in T-cell leukemia are still poorly understood. We identify here a specific novel property of Notch3 that is acetylated and deacetylated at lysines 1692 and 1731 by p300 and HDAC1, respectively, a balance impaired by HDAC inhibitors (HDACi) that favor hyperacetylation. By using HDACi and a non-acetylatable Notch3 mutant carrying K/R1692−1731 mutations in the intracellular domain, we show that Notch3 acetylation primes ubiquitination and proteasomal-mediated degradation of the protein. As a consequence, Notch3 protein expression and its transcriptional activity are decreased both in vitro and in vivo in Notch3 transgenic (tg) mice, thus impairing downstream signaling upon target genes. Consistently, Notch3-induced T-cell proliferation is inhibited by HDACi, whereas it is enhanced by the non-acetylatable Notch3-K/R1692−1731 mutant. Finally, HDACi-induced Notch3 hyperacetylation prevents in vivo growth of T-cell leukemia/lymphoma in Notch3 tg mice. Together, our findings suggest a novel level of Notch signaling control in which Notch3 acetylation/deacetylation process represents a key regulatory switch, thus representing a suitable druggable target for Notch3-sustained T-cell acute lymphoblastic leukemia therapy.


Oncogene | 2016

Prolyl-isomerase Pin1 controls Notch3 protein expression and regulates T-ALL progression

G. Franciosa; G. Diluvio; F Del Gaudio; M.V. Giuli; Rocco Palermo; Paola Grazioli; Antonio Francesco Campese; Claudio Talora; Diana Bellavia; G D'Amati; Zein Mersini Besharat; C Nicoletti; C W Siebel; L Choy; Alessandra Rustighi; G Del Sal; Isabella Screpanti; Saula Checquolo

Deregulated Notch signaling is associated with T-cell Acute Lymphoblastic Leukemia (T-ALL) development and progression. Increasing evidence reveals that Notch pathway has an important role in the invasion ability of tumor cells, including leukemia, although the underlying molecular mechanisms remain mostly unclear. Here, we show that Notch3 is a novel target protein of the prolyl-isomerase Pin1, which is able to regulate Notch3 protein processing and to stabilize the cleaved product, leading to the increased expression of the intracellular domain (N3IC), finally enhancing Notch3-dependent invasiveness properties. We demonstrate that the combined inhibition of Notch3 and Pin1 in the Notch3-overexpressing human leukemic TALL-1 cells reduces their high invasive potential, by decreasing the expression of the matrix metalloprotease MMP9. Consistently, Pin1 depletion in a mouse model of Notch3-induced T-ALL, by reducing N3IC expression and signaling, impairs the expansion/invasiveness of CD4+CD8+ DP cells in peripheral lymphoid and non-lymphoid organs. Notably, in in silico gene expression analysis of human T-ALL samples we observed a significant correlation between Pin1 and Notch3 expression levels, which may further suggest a key role of the newly identified Notch3-Pin1 axis in T-ALL aggressiveness and progression. Thus, combined suppression of Pin1 and Notch3 proteins may be exploited as an additional target therapy for T-ALL.


Leukemia | 2013

Glucocorticoid sensitivity of T-cell lymphoblastic leukemia/lymphoma is associated with glucocorticoid receptor-mediated inhibition of Notch1 expression

Samantha Cialfi; Rocco Palermo; Sonia Manca; Saula Checquolo; Diana Bellavia; Maria Pelullo; Roberta Quaranta; Carlo Dominici; Alberto Gulino; Isabella Screpanti; Claudio Talora

Glucocorticoid sensitivity of T-cell lymphoblastic leukemia/lymphoma is associated with glucocorticoid receptor-mediated inhibition of Notch1 expression

Collaboration


Dive into the Saula Checquolo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Diana Bellavia

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Alberto Gulino

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Rocco Palermo

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claudio Talora

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Luigi Frati

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Maria Pia Felli

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Paola Grazioli

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Alessandra Vacca

Sapienza University of Rome

View shared research outputs
Researchain Logo
Decentralizing Knowledge