Network


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

Hotspot


Dive into the research topics where Chiara Novello is active.

Publication


Featured researches published by Chiara Novello.


International Journal of Oncology | 2013

miRNA expression profile in human osteosarcoma: role of miR-1 and miR-133b in proliferation and cell cycle control.

Chiara Novello; Laura Pazzaglia; Chiara Cingolani; Amalia Conti; Irene Quattrini; Maria Cristina Manara; Mauro Tognon; Piero Picci; Maria Serena Benassi

miRNA profile deregulation affecting downstream signaling pathways activates endpoints that represent potential biomarkers for prognosis and treatment of tumor patients. In the past 20 years conventional therapy for osteosarcoma (OS) reached a survival plateau, highlighting the need for new therapeutic approaches. In this study, microarray unsupervised and supervised analysis identified, respectively, 100 and 40 differentially expressed miRNAs in OS samples with different grades of malignancy compared to normal bone. When analyzing low-grade and high-grade OS by unsupervised analysis, 12 miRNAs were found to be differentially expressed. Real‑time PCR performed on a larger series of OS confirmed a significant lower expression of miR-1, miR‑133b and miR-378* in tumors with respect to control, also showing lower mRNA levels in 31 high-grade OS than in 25 low-grade and in metastatic versus non‑metastatic patients. We demonstrated that miR-1 and miR133b were downregulated in OS cell lines compared to normal osteoblasts. Secondly, by transfection with miRNA precursor molecules, we demonstrated that the ectopic expression of miR-1 and miR-133b in U2-OS cell lines significantly reduced cell proliferation and MET protein expression and negatively regulated cell invasiveness and motility in a short-term assay. Cell cycle distribution revealed block in G(1) and delay of cell cycle progression associated with increased apoptosis in miR-1- and miR‑133b‑transfected cells, respectively. Our data assessed specific miRNA profiling deregulation in OS clinical samples and suggest that the expression of miR-1 and miR-133b may control cell proliferation and cell cycle through MET protein expression modulation.


American Journal of Pathology | 2011

Identification of Potential Biomarkers for Giant Cell Tumor of Bone Using Comparative Proteomics Analysis

Amalia Conti; Gemma Caballero Rodriguez; Antonella Chiechi; Rosa Maria Dégano Blazquez; Victoria Barbado; Tibor Krenács; Chiara Novello; Laura Pazzaglia; Irene Quattrini; Licciana Zanella; Piero Picci; Enrique de Alava; Maria Serena Benassi

Giant cell tumor of bone can be locally aggressive and occasionally can metastasize in the lungs. To identify new markers predictive of aggressive behavior, we analyzed five patients who developed lung metastasis and five who remained disease free for a minimum of 5 years. Using two-dimensional electrophoresis, we detected 28 differentially expressed spots. Fourteen spots were identified using mass spectrometry, including seven up-regulated and seven down-regulated in metastatic samples and classified according to functional categories. We then selected five proteins involved in cell cycle or apoptosis. Thioredoxin peroxidase, allograft inflammatory factor 1, and ubiquitin E2N had more than threefold up-regulation; glutathione peroxidase 1 had 1.9-fold up-regulation; and heat shock protein 27 showed down-regulation in metastatic samples with a very low P value. After validation and analysis of protein levels, evaluation of clinical impact was assessed in a much wider cohort of primary archival specimens. Immunodetection showed a higher frequency of thioredoxin peroxidase, allograft inflammatory factor 1, ubiquitin E2N, and glutathione peroxidase 1 overexpression in primary tumors that developed into lung metastases or that locally relapsed than in the disease-free group, with variable stain intensity and distribution. Kaplan-Meier analysis showed that high expression of glutathione peroxidase 1 was strongly related to local recurrence and metastasis, suggesting that its up-regulation may identify a subset of high-risk patients with giant cell tumor prone to receive diverse clinical management.


Cellular Oncology | 2012

MicroRNA cloning and sequencing in osteosarcoma cell lines: differential role of miR-93

Luisa Montanini; Lisa Lasagna; Valeria Barili; Søren Peter Jonstrup; Alba Murgia; Laura Pazzaglia; Amalia Conti; Chiara Novello; Jørgen Kjems; Roberto Perris; Maria Serena Benassi

BackgroundStudies show that abnormalities in non-coding genes can contribute to carcinogenesis; microRNA levels may modulate cancer growth and metastatic diffusion.MethodMicroRNA libraries were built and sequenced from two osteosarcoma cell lines (MG-63 and 143B), which differ in proliferation and transmigration. By cloning and transfection, miR-93, expressed in both cell lines, was then investigated for its involvement in osteosarcoma progression.ResultsSix of the 19 miRNA identified were expressed in both cell lines with higher expression levels of miR-93 in 143B and in primary osteosarcoma cultures compared to normal osteoblasts. Interestingly, levels of miR-93 were significantly higher in metastases from osteosarcoma than in paired primary tumours. When 143B and MG-63 were transfected with miR-93, clones appeared to respond differently to microRNA overexpression. Ectopic expression of miR-93 more significantly increased cell proliferation and invasivity in 143B than in MG-63 clones. Furthermore, increased mRNA and protein levels of E2F1, one of the potential miR-93 targets, were seen in osteosarcoma cellular clones and its involvement in 143B cell proliferation was confirmed by E2F1 silencing.ConclusionAlthough further studies are needed to evaluate miRNA involvement in osteosarcoma progression, miR-93 overexpression seems to play an important role in osteosarcoma cell growth and invasion.


Clinical Cancer Research | 2013

Elevated TNFR1 and Serotonin in Bone Metastasis Are Correlated with Poor Survival following Bone Metastasis Diagnosis for Both Carcinoma and Sarcoma Primary Tumors

Antonella Chiechi; Chiara Novello; Giovanna Magagnoli; Emanuel F. Petricoin; Jianghong Deng; Maria Serena Benassi; Piero Picci; Iosif I. Vaisman; Virginia Espina; Lance A. Liotta

Purpose: There is an urgent need for therapies that will reduce the mortality of patients with bone metastasis. In this study, we profiled the protein signal pathway networks of the human bone metastasis microenvironment. The goal was to identify sets of interacting proteins that correlate with survival time following the first diagnosis of bone metastasis. Experimental Design: Using Reverse Phase Protein Microarray technology, we measured the expression of 88 end points in the bone microenvironment of 159 bone metastasis tissue samples derived from patients with primary carcinomas and sarcomas. Results: Metastases originating from different primary tumors showed similar levels of cell signaling across tissue types for the majority of proteins analyzed, suggesting that the bone microenvironment strongly influences the metastatic tumor signaling profiles. In a training set (72 samples), TNF receptor 1, alone (P = 0.0013) or combined with serotonin (P = 0.0004), TNFα (P = 0.0214), and RANK (P = 0.0226), was associated with poor survival, regardless of the primary tumor of origin. Results were confirmed by (i) analysis of an independent validation set (71 samples) and (ii) independent bioinformatic analysis using a support vector machine learning model. Spearman rho analysis revealed a highly significant number of interactions intersecting with ERα S118, serotonin, TNFα, RANKL, and matrix metalloproteinase in the bone metastasis signaling network, regardless of the primary tumor. The interaction network pattern was significantly different in the short versus long survivors. Conclusions: TNF receptor 1 and neuroendocrine-regulated protein signal pathways seem to play an important role in bone metastasis and may constitute a novel drug-targetable mechanism of seed-soil cross talk in bone metastasis. Clin Cancer Res; 19(9); 2473–85. ©2013 AACR.


Genes, Chromosomes and Cancer | 2014

Differentiating soft tissue leiomyosarcoma and undifferentiated pleomorphic sarcoma: A miRNA analysis.

Mohamed Guled; Laura Pazzaglia; Ioana Borze; Neda Mosakhani; Chiara Novello; Maria Serena Benassi; Sakari Knuutila

The rare and highly aggressive adult soft tissue sarcomas leiomyosarcoma (LMS) and undifferentiated pleomorphic sarcoma (UPS) contain complex genomes characterized by a multitude of rearrangements, amplifications, and deletions. Differential diagnosis remains a challenge. MicroRNA (miRNA) profiling was conducted on a series of LMS and UPS samples to initially investigate the differential expression and to identify specific signatures useful for improving the differential diagnosis. Initially, 10 high‐grade LMS and 10 high‐grade UPS were profiled with a miRNA microarray. Two cultured human mesenchymal stem cell samples were used as controls. 38 and 46 miRNAs classified UPS and LMS samples, respectively, into separate groups compared to control samples. When comparing the two profiles, miR‐199b‐5p, miR‐320a, miR‐199a‐3p, miR‐126, miR‐22 were differentially expressed. These were validated by RT‐PCR on a further series of 27 UPS and 21 LMS for a total of 68 cases. The levels of miR‐199‐5p and miR‐320a, in particular, confirmed the microarray data, the former highly expressed in UPS and the latter in LMS. Immunohistochemistry was performed on all 68 cases to confirm original diagnosis. Recently reported LMS‐ and UPS‐associated genes were correlated with miRNA targets based on target algorithms of three databases. Several genes including IMP3, ROR2, MDM2, CDK4, and UPA, are targets of differentially expressed miRNAs. We identified miRNA expression patterns in LMS and UPS, linking them to chromosomal regions and mRNA targets known to be involved in tumor development/progression of LMS and UPS.


PLOS ONE | 2014

p53-dependent activation of microRNA-34a in response to etoposide-induced DNA damage in osteosarcoma cell lines not impaired by dominant negative p53 expression.

Chiara Novello; Laura Pazzaglia; Amalia Conti; Irene Quattrini; Paola Perego; Piero Picci; Maria Serena Benassi

Osteosarcoma (OS) is the most common primary malignant bone tumor and prevalently occurs in the second decade of life. Etoposide, a chemotherapeutic agent used in combined treatments of recurrent human OS, belongs to the topoisomerase inhibitor family and causes DNA breakage. In this study we evaluated the cascade of events determined by etoposide-induced DNA damage in OS cell lines with different p53 status focusing on methylation status and expression of miR-34a that modulate tumor cell growth and cell cycle progression. Wild-type p53 U2-OS cells and U2-OS cells expressing dominant-negative form of p53 (U2- OS175) were more sensitive to etoposide than p53-deficient MG63 and Saos-2 cells, showing increased levels of unmethylated miR-34a, reduced expression of CDK4 and cell cycle arrest in G1 phase. In contrast, MG63 and Saos-2 cell lines presented aberrant methylation of miR-34a promoter gene with no miR-34a induction after etoposide treatment, underlining the close connection between p53 expression and miR-34a methylation status. Consistently, in p53siRNA transfected U2-OS cells we observed loss of miR-34a induction after etoposide exposure associated with a partial gain of gene methylation and cell cycle progress towards G2/M phase. Our results suggest that the open and unmethylated conformation of the miR-34a gene may be regulated by p53 able to bind the gene promoter. In conclusion, cell response to etoposide-induced DNA damage was not compromised in cells with dominant-negative p53 expression.


Clinical & Experimental Metastasis | 2014

Mapping protein signal pathway interaction in sarcoma bone metastasis: linkage between rank, metalloproteinases turnover and growth factor signaling pathways.

Amalia Conti; Virginia Espina; Antonella Chiechi; Giovanna Magagnoli; Chiara Novello; Laura Pazzaglia; Irene Quattrini; Piero Picci; Lance A. Liotta; Maria Serena Benassi

We applied reverse phase protein microarrays technology to map signal pathway interactions in a discovery set of 34 soft tissue sarcoma (STS) bone metastases compared to healthy bone. Proteins associated with matrix remodeling (MMP), adhesion (FAK Y576/577, Syndecan-1), and growth/survival (IGF1R Y1135/1136, PI3K, EGFR) were elevated in metastasis compared to normal bone. Linkage between Syndecan-1, FAK Y576/577, Shc Y317, and EGFR, IGF Y1135/1136, PI3K/AKT was a prominent feature of STS bone metastasis. Elevated linkage between RANKL and 4EBP1 T37/46, EGFR, IGF1R Y1135/1136, Src Y41, Shc Y317, PI3Kp110γ was associated with short survival. Finally, we tested the hypothesis that signal pathway proteins augmented in the STS bone metastasis may provide clues to understand the subset of primary STS that metastasize. The most representative molecules identified in the discovery set were validated on an independent series of 82 primary STS by immunohistochemistry applied to a tissue microarray. The goal was to correlate the molecular profile in the primary tumors with a higher likelihood of metastasis. Elevation of activated kinase substrate endpoints IRS1 S612, 4EBP1 T37/46, FAK Y576/577 and loss of Fibronectin, were associated with a higher likelihood of metastases. These data indicate that the linkage between matrix remodeling, adhesion, and growth signaling may drive STS metastasis and can be the basis for prognostic and therapeutic strategies.


Oncology Reports | 2014

Metformin inhibits growth and sensitizes osteosarcoma cell lines to cisplatin through cell cycle modulation

Irene Quattrini; Amalia Conti; Laura Pazzaglia; Chiara Novello; Stefano Ferrari; Piero Picci; Maria Serena Benassi

Osteosarcoma (OS) is the most common cancer that affects the bone and appears to be resistant to several chemotherapeutic drugs. The aim of the present study was to verify whether the combination of metformin and cisplatin has an effect on OS cell lines. OS cell lines U2OS, 143B and MG63 were treated with metformin, cisplatin or a combination of both drugs. Viability, apoptosis and cell cycle were evaluated to characterize the effects of the treatments. Western blot analyses were used to evaluate protein expression. All OS cell lines were found to be sensitive to metformin with different values of IC50, showing a slowdown of cell cycle associated or not with apoptosis. In particular, metformin was able to sensitize cells to cisplatin, to which all OS cell lines were resistant, demonstrating a synergistic effect in the combined treatment of the two drugs. The data obtained may have clinical relevance for novel therapeutic strategies for the treatment of OS; metformin inhibits tumor cell growth and amplifies the effect of cisplatin.


Journal of Orthopaedic Research | 2015

Prognostic role of nuclear factor/IB and bone remodeling proteins in metastatic giant cell tumor of bone: A retrospective study

Irene Quattrini; Laura Pazzaglia; Amalia Conti; Chiara Novello; Cristina Ferrari; Elettra Pignotti; Piero Picci; Maria Serena Benassi

Giant cell tumor of bone (GCTb) represents 5% of bone tumors, and although considered benign, 5% metastasize to the lung. The expression of proteins directly or indirectly associated with osteolysis and tumor growth was studied on 163 samples of GCTb. Of these, 33 patients developed lung metastasis during follow‐up. The impact of tumor–host interaction on clinical aspects was evaluated with the aim of finding specific markers for new biological therapies, thus improving clinical management of GCTb. Protein expression was evaluated by immunohistochemical analysis on Tissue Microarray. The majority of GCTb samples from patients with metastatic disease were strongly positive to RANKL and its receptor RANK as well as to CAII and MMP‐2 and to pro‐survival proteins NFIB and c‐Fos. Kaplan–Meier analysis indicated a significant difference in metastasis free survival curves based on protein staining. Interestingly, the statistical correlation established a strong association between all variables studied with a higher τ coefficient for RANK/RANKL, RANK/NFIB, and RANKL/NFIB pairs. At multivariate analysis co‐overexpression of NFIB, RANK and RANKL significantly increased the risk of metastasis with an odds ratio of 13.59 (95%CI 4.12–44.82; p < 0.0005). In conclusion, the interconnection between matrix remodeling and tumor cell activity may identify tumor–host endpoints for new biological treatments.


Hepatology | 2017

A Phosphokinome-based screen uncovers new drug synergies for cancer driven by liver-specific gain of non-oncogenic RTKs.

Yannan Fan; Maria Arechederra; Sylvie Richelme; Fabrice Daian; Chiara Novello; Julien Calderaro; Luca Di Tommaso; Guillaume Morcrette; Sandra Rebouissou; Matteo Donadon; Emanuela Morenghi; Jessica Zucman-Rossi; Massimo Roncalli; Rosanna Dono; Flavio Maina

Genetic mutations leading to oncogenic variants of receptor tyrosine kinases (RTKs) are frequent events during tumorigenesis; however, the cellular vulnerability to nononcogenic RTK fluctuations has not been characterized. Here, we demonstrated genetically that in the liver subtle increases in wild‐type Met RTK levels are sufficient for spontaneous tumors in mice (Alb‐R26Met), conceptually illustrating how the shift from physiological to pathological conditions results from slight perturbations in signaling dosage. By analyzing 96 different genes in a panel of tumor samples, we demonstrated that liver tumorigenesis modeled by Alb‐R26Met mice corresponds to a subset of hepatocellular carcinoma (HCC) patients, thus establishing the clinical relevance of this HCC mouse model. We elucidated the regulatory networks underlying tumorigenesis by combining a phosphokinome screen with bioinformatics analysis. We then used the signaling diversity results obtained from Alb‐R26Met HCC versus control livers to design an “educated guess” drug screen, which led to the identification of new, deleterious synthetic lethal interactions. In particular, we report synergistic effects of mitogen‐activated protein kinase kinase, ribosomal S6 kinase, and cyclin‐dependent kinase 1/2 in combination with Bcl‐XL inhibition on a panel of liver cancer cells. Focusing on mitogen‐activated protein kinase kinase and Bcl‐XL targeting, we mechanistically demonstrated concomitant down‐regulation of phosphorylated extracellular signal–regulated kinase and myeloid cell leukemia 1 levels. Of note, a phosphorylated extracellular signal–regulated kinase+/BCL‐XL+/myeloid cell leukemia 1+ signature, deregulated in Alb‐R26Met tumors, characterizes a subgroup of HCC patients with poor prognosis. Conclusion: Our genetic studies highlight the heightened vulnerability of liver cells to subtle changes in nononcogenic RTK levels, allowing them to acquire a molecular profile that facilitates the full tumorigenic program; furthermore, our outcomes uncover new synthetic lethal interactions as potential therapies for a cluster of HCC patients. (Hepatology 2017;66:1644–1661).

Collaboration


Dive into the Chiara Novello's collaboration.

Top Co-Authors

Avatar

Amalia Conti

George Mason University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge