Network


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

Hotspot


Dive into the research topics where Cristina Sorino is active.

Publication


Featured researches published by Cristina Sorino.


Cancer Cell | 2010

Che-1 Promotes Tumor Cell Survival by Sustaining Mutant p53 Transcription and Inhibiting DNA Damage Response Activation

Tiziana Bruno; Agata Desantis; Gianluca Bossi; Silvia Di Agostino; Cristina Sorino; Francesca De Nicola; Simona Iezzi; Annapaola Franchitto; Barbara Benassi; Sergio Galanti; Francesca La Rosa; Aristide Floridi; Alfonso Bellacosa; Claudio Passananti; Giovanni Blandino; Maurizio Fanciulli

Che-1 is a RNA polymerase II binding protein involved in the regulation of gene transcription and, in response to DNA damage, promotes p53 transcription. In this study, we investigated whether Che-1 regulates mutant p53 expression. We found that Che-1 is required for sustaining mutant p53 expression in several cancer cell lines, and that Che-1 depletion by siRNA induces apoptosis both in vitro and in vivo. Notably, loss of Che-1 activates DNA damage checkpoint response and induces transactivation of p73. Therefore, these findings underline the important role that Che-1 has in survival of cells expressing mutant p53.


The EMBO Journal | 2015

Che-1-induced inhibition of mTOR pathway enables stress-induced autophagy

Agata Desantis; Tiziana Bruno; Valeria Catena; Francesca De Nicola; Frauke Goeman; Simona Iezzi; Cristina Sorino; Maurilio Ponzoni; Gianluca Bossi; Vincenzo Federico; Francesca La Rosa; Maria Rosaria Ricciardi; Elena Lesma; Paolo D'Onorio De Meo; Tiziana Castrignanò; Maria Teresa Petrucci; Francesco Pisani; Marta Chesi; P. Leif Bergsagel; Aristide Floridi; Giovanni Tonon; Claudio Passananti; Giovanni Blandino; Maurizio Fanciulli

Mammalian target of rapamycin (mTOR) is a key protein kinase that regulates cell growth, metabolism, and autophagy to maintain cellular homeostasis. Its activity is inhibited by adverse conditions, including nutrient limitation, hypoxia, and DNA damage. In this study, we demonstrate that Che‐1, a RNA polymerase II‐binding protein activated by the DNA damage response, inhibits mTOR activity in response to stress conditions. We found that, under stress, Che‐1 induces the expression of two important mTOR inhibitors, Redd1 and Deptor, and that this activity is required for sustaining stress‐induced autophagy. Strikingly, Che‐1 expression correlates with the progression of multiple myeloma and is required for cell growth and survival, a malignancy characterized by high autophagy response.


Cell Death and Disease | 2015

Che-1 modulates the decision between cell cycle arrest and apoptosis by its binding to p53

Agata Desantis; Tiziana Bruno; Valeria Catena; F De Nicola; Frauke Goeman; Simona Iezzi; Cristina Sorino; M P Gentileschi; S Germoni; V Monteleone; M Pellegrino; M Kann; P D De Meo; Matteo Pallocca; Katja Höpker; Francesca Moretti; Elisabetta Mattei; H C Reinhardt; Aristide Floridi; Claudio Passananti; Thomas Benzing; Giovanni Blandino; Maurizio Fanciulli

The tumor suppressor p53 is mainly involved in the transcriptional regulation of a large number of growth-arrest- and apoptosis-related genes. However, a clear understanding of which factor/s influences the choice between these two opposing p53-dependent outcomes remains largely elusive. We have previously described that in response to DNA damage, the RNA polymerase II-binding protein Che-1/AATF transcriptionally activates p53. Here, we show that Che-1 binds directly to p53. This interaction essentially occurs in the first hours of DNA damage, whereas it is lost when cells undergo apoptosis in response to posttranscriptional modifications. Moreover, Che-1 sits in a ternary complex with p53 and the oncosuppressor Brca1. Accordingly, our analysis of genome-wide chromatin occupancy by p53 revealed that p53/Che1 interaction results in preferential transactivation of growth arrest p53 target genes over its pro-apoptotic target genes. Notably, exposure of Che-1+/− mice to ionizing radiations resulted in enhanced apoptosis of thymocytes, compared with WT mice. These results confirm Che-1 as an important regulator of p53 activity and suggest Che-1 to be a promising yet attractive drug target for cancer therapy.


Journal of Biological Chemistry | 2013

Centrosomal Che-1 Protein Is Involved in the Regulation of Mitosis and DNA Damage Response by Mediating Pericentrin (PCNT)-dependent Chk1 Protein Localization

Cristina Sorino; Tiziana Bruno; Agata Desantis; Maria Grazia Di Certo; Simona Iezzi; Francesca De Nicola; Valeria Catena; Aristide Floridi; Luciana Chessa; Claudio Passananti; Enrico Cundari; Maurizio Fanciulli

Background: Che-1 is an RNA polymerase II-binding protein involved in gene transcription, cell proliferation, and DNA damage response. Results: Che-1 localizes at interphase centrosomes. Che-1 inhibition abolishes Chk1 localization at centrosomes, advancing entry into mitosis. Conclusion: Che-1 acts like an upstream regulator of Chk1 centrosomal functions. Significance: Che-1 inhibition might potentiate tumor cell sensitivity to antimitotic drugs. To combat threats posed by DNA damage, cells have evolved mechanisms, collectively termed DNA damage response (DDR). These mechanisms detect DNA lesions, signal their presence, and promote their repair. Centrosomes integrate G2/M checkpoint control and repair signals in response to genotoxic stress, acting as an efficient control mechanism when G2/M checkpoint function fails and mitosis begins in the presence of damaged DNA. Che-1 is an RNA polymerase II-binding protein involved in the regulation of gene transcription, induction of cell proliferation, and DDR. Here we provide evidence that in addition to its nuclear localization, Che-1 localizes at interphase centrosomes, where it accumulates following DNA damage or spindle poisons. We show that Che-1 depletion generates supernumerary centrosomes, multinucleated cells, and multipolar spindle formation. Notably, Che-1 depletion abolishes the ability of Chk1 to bind pericentrin and to localize at centrosomes, which, in its turn, deregulates the activation of centrosomal cyclin B-Cdk1 and advances entry into mitosis. Our results reinforce the notion that Che-1 plays an important role in DDR and that its contribution seems to be relevant for the spindle assembly checkpoint.


Cell Death and Disease | 2014

HIPK2 sustains apoptotic response by phosphorylating Che-1/AATF and promoting its degradation

F De Nicola; Valeria Catena; C Rinaldo; Tiziana Bruno; Simona Iezzi; Cristina Sorino; Agata Desantis; S Camerini; Marco Crescenzi; Aristide Floridi; Claudio Passananti; Silvia Soddu; Maurizio Fanciulli

Che-1/AATF is an RNA polymerase II-binding protein that is involved in the regulation of gene transcription, which undergoes stabilization and accumulation in response to DNA damage. We have previously demonstrated that following apoptotic induction, Che-1 protein levels are downregulated through its interaction with the E3 ligase HDM2, which leads to Che-1 degradation by ubiquitylation. This interaction is mediated by Pin1, which determines a phosphorylation-dependent conformational change. Here we demonstrate that HIPK2, a proapoptotic kinase, is involved in Che-1 degradation. HIPK2 interacts with Che-1 and, upon genotoxic stress, phosphorylates it at specific residues. This event strongly increases HDM2/Che-1 interaction and degradation of Che-1 protein via ubiquitin-dependent proteasomal system. In agreement with these findings, we found that HIPK2 depletion strongly decreases Che-1 ubiquitylation and degradation. Notably, Che-1 overexpression strongly counteracts HIPK2-induced apoptosis. Our results establish Che-1 as a new HIPK2 target and confirm its important role in the cellular response to DNA damage.


Oncotarget | 2016

Deptor transcriptionally regulates endoplasmic reticulum homeostasis in multiple myeloma cells

Valeria Catena; Tiziana Bruno; Francesca De Nicola; Frauke Goeman; Matteo Pallocca; Simona Iezzi; Cristina Sorino; Giovanni Cigliana; Aristide Floridi; Giovanni Blandino; Maurizio Fanciulli

Multiple myeloma (MM) is a malignant disorder of plasma cells characterized by active production and secretion of monoclonal immunoglobulins (IgG), thus rendering cells prone to endoplasmic reticulum (ER) stress. For this reason, MM cell survival requires to maintain ER homeostasis at basal levels. Deptor is an mTOR binding protein, belonging to the mTORC1 and mTORC2 complexes. It was reported that Deptor is overexpressed in MM cells where it inhibits mTOR kinase activity and promotes cell survival by activating Akt signaling. Here we identify Deptor as a nuclear protein, able to bind DNA and regulate transcription in MM cells. In particular, we found that Deptor plays an important role in the maintenance of the ER network, sustaining the expression of several genes involved in this pathway. In agreement with this, Deptor depletion induces ER stress and synergizes the effect of the proteasome inhibitor bortezomib (Bz) in MM cells. These findings provide important new insights in the ER stress control in MM cells.


Journal of Experimental & Clinical Cancer Research | 2017

Che-1 sustains hypoxic response of colorectal cancer cells by affecting Hif-1α stabilization

Tiziana Bruno; Mariacristina Valerio; Luca Casadei; Francesca De Nicola; Frauke Goeman; Matteo Pallocca; Valeria Catena; Simona Iezzi; Cristina Sorino; Agata Desantis; Cesare Manetti; Giovanni Blandino; Aristide Floridi; Maurizio Fanciulli

BackgroundSolid tumours are less oxygenated than normal tissues. Consequently, cancer cells acquire to be adapted to a hypoxic environment. The poor oxygenation of solid tumours is also a major indicator of an adverse cancer prognosis and leads to resistance to conventional anticancer treatments. We previously showed the involvement of Che-1/AATF (Che-1) in cancer cell survival under stress conditions. Herein we hypothesized that Che-1 plays a role in the response of cancer cells to hypoxia.MethodsThe human colon adenocarcinoma HCT116 and HT29 cell lines undepleted or depleted for Che-1 expression by siRNA, were treated under normoxic and hypoxic conditions to perform studies regarding the role of this protein in metabolic adaptation and cell proliferation. Che-1 expression was detected using western blot assays; cell metabolism was assessed by NMR spectroscopy and functional assays. Additional molecular studies were performed by RNA seq, qRT-PCR and ChIP analyses.ResultsHere we report that Che-1 expression is required for the adaptation of cells to hypoxia, playing an important role in metabolic modulation. Indeed, Che-1 depletion impacted on HIF-1α stabilization, thus downregulating the expression of several genes involved in the response to hypoxia and affecting glucose metabolism.ConclusionsWe show that Che-1 a novel player in the regulation of HIF-1α in response to hypoxia. Notably, we found that Che-1 is required for SIAH-2 expression, a member of E3 ubiquitin ligase family that is involved in the degradation of the hydroxylase PHD3, the master regulator of HIF-1α stability.


EMBO Reports | 2018

Che‐1 is targeted by c‐Myc to sustain proliferation in pre‐B‐cell acute lymphoblastic leukemia

Valentina Folgiero; Cristina Sorino; Matteo Pallocca; Francesca De Nicola; Frauke Goeman; Valentina Bertaina; Luisa Strocchio; Paolo Romania; Angela Pitisci; Simona Iezzi; Valeria Catena; Tiziana Bruno; Georgios Strimpakos; Claudio Passananti; Elisabetta Mattei; Giovanni Blandino; Franco Locatelli; Maurizio Fanciulli

Despite progress in treating B‐cell precursor acute lymphoblastic leukemia (BCP‐ALL), disease recurrence remains the main cause of treatment failure. New strategies to improve therapeutic outcomes are needed, particularly in high‐risk relapsed patients. Che‐1/AATF (Che‐1) is an RNA polymerase II‐binding protein involved in proliferation and tumor survival, but its role in hematological malignancies has not been clarified. Here, we show that Che‐1 is overexpressed in pediatric BCP‐ALL during disease onset and at relapse, and that its depletion inhibits the proliferation of BCP‐ALL cells. Furthermore, we report that c‐Myc regulates Che‐1 expression by direct binding to its promoter and describe a strict correlation between Che‐1 expression and c‐Myc expression. RNA‐seq analyses upon Che‐1 or c‐Myc depletion reveal a strong overlap of the respective controlled pathways. Genomewide ChIP‐seq experiments suggest that Che‐1 acts as a downstream effector of c‐Myc. These results identify the pivotal role of Che‐1 in the control of BCP‐ALL proliferation and present the protein as a possible therapeutic target in children with relapsed BCP‐ALL.


Journal of Experimental & Clinical Cancer Research | 2018

Recent advances in searching c-Myc transcriptional cofactors during tumorigenesis

Matteo Caforio; Cristina Sorino; Stefano Iacovelli; Maurizio Fanciulli; Franco Locatelli; Valentina Folgiero

BackgroundThe mechanism by which c-Myc exerts its oncogenic functions is not completely clear and different hypotheses are still under investigation. The knowledge of the capacity of c-Myc to bind exclusively E-box sequences determined the discrepancy between, on the one hand, genomic studies showing the binding of c-Myc to all active promoters and, on the other hand, the evidence that only 60% or less of the binding sites have E-box sequences.Main bodyIn this review, we provide support to the hypothesis that the cooperation of c-Myc with transcriptional cofactors mediates c-Myc-induced cellular functions. We produce evidence that recently identified cofactors are involved in c-Myc control of survival mechanisms of cancer cells.ConclusionThe identification of new c-Myc cofactors could favor the development of therapeutic strategies able to compensate the difficulty of targeting c-Myc.


Cell Cycle | 2018

A new baby in the c-Myc-directed transcriptional machinery: Che-1/AATF

Valentina Folgiero; Cristina Sorino; Franco Locatelli; Maurizio Fanciulli

ABSTRACT B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is the most common malignancy in childhood. Despite the high cure-rate, identifying new druggable molecular targets is still of great interest. In a cohort of BCP-ALL pediatric patients, irrespectively of the molecule/karyotype lesions found, we recently observed high expression of c-Myc and Che-1/AATF, which disappears at time of remission. Study of the molecular mechanisms involved in this co-expression revealed that Che-1 expression was crucial for induction of blast-cell proliferation driven by c-Myc. Furthermore, Che-1/AATF silencing in primary BCP-ALL cell lines improves responsiveness to chemotherapy. These data individuate Che-1 as a possible novel target in the treatment of BCP-ALL able to affect c-Myc-driven tumorigenicity.

Collaboration


Dive into the Cristina Sorino's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claudio Passananti

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Simona Iezzi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gianluca Bossi

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge