Network


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

Hotspot


Dive into the research topics where Raffaela Barbano is active.

Publication


Featured researches published by Raffaela Barbano.


Epigenetics | 2011

Frequent epigenetics inactivation of KEAP1 gene in non-small cell lung cancer

Lucia Anna Muscarella; Paola Parrella; Vito D’Alessandro; Annamaria la Torre; Raffaela Barbano; Andrea Fontana; Antonio Tancredi; Vito Guarnieri; Teresa Balsamo; Michelina Coco; Massimiliano Copetti; Fabio Pellegrini; Patrizia De Bonis; Michele Bisceglia; Gerardo Scaramuzzi; Evaristo Maiello; Vanna Maria Valori; Giuseppe Merla; Gianluigi Vendemiale; Vito Michele Fazio

The KEAP1/Nrf2 pathway is a master regulator of several redox-sensitive genes implicated in resistance of tumor cells against chemotherapeutic drugs. Recent data suggest that epigenetic mechanisms may play a pivotal role in the regulation of KEAP1 expression. We performed a comprehensive genetic and epigenetic analysis of the KEAP1 gene in 47 non-small cell lung cancer tissues and normal specimens. Promoter methylation analysis was performed using a quantitative methylation specific PCR assay in real time. Methylation at the KEAP1 promoter region was detected in 22 out of the 47 NSCLCs (47%) and in none of the normal tissues analyzed. Somatic mutations were detected in 7 out of the 47 tumors (15%) and loss of heterozygosity (LOH) in 10 out of the 47 (21%) of the cases. Overall, we found at least one molecular alteration in 57% of the cases. Approximately one third of the tumors had two alterations and this feature was associated with higher risk of disease progression in univariate COX regression analysis (HR = 3.62; 95% CI 1.24–10.65, p = 0.02). This result was confirmed by Kaplan-Meier analysis, which demonstrated an association between worst outcome and KEAP1 double alterations (p = 0.01, Log rank test). Our results further suggest that deregulation of the NRF2/KEAP1 system could play a pivotal role in the cancerogenesis of NSCLC. In addition identifying patients with KEAP1 genetic and epigenetic abnormalities may contribute to disease progression prediction and response to therapy in lung cancer patients.


Cancer Epidemiology, Biomarkers & Prevention | 2009

Changes in CpG Islands Promoter Methylation Patterns during Ductal Breast Carcinoma Progression

Mohammad O. Hoque; Maria Prencipe; Maria Luana Poeta; Raffaela Barbano; Vanna Maria Valori; Massimiliano Copetti; A. Gallo; Mariana Brait; Evaristo Maiello; Adolfo Apicella; Raffaele Rossiello; Francesco Zito; Tommasi Stefania; Angelo Paradiso; Massimo Carella; Bruno Dallapiccola; Roberto Murgo; Illuminato Carosi; Michele Bisceglia; Vito Michele Fazio; David Sidransky; Paola Parrella

Aberrant promoter methylation of several known or putative tumor suppressor genes occurs frequently during carcinogenesis, and this epigenetic change has been considered as a potential molecular marker for cancer. We examined the methylation status of nine genes (APC, CDH1, CTNNB1, TIMP3, ESR1, GSTP1, MGMT, THBS1, and TMS1), by quantitative methylation specific PCR. Synchronous preinvasive lesions (atypical ductal hyperplasia and/or ductal carcinoma in situ) and invasive ductal breast carcinoma from 52 patients, together with pure lesions from 24 patients and 12 normal tissues paired to tumor and 20 normal breast distant from tumor were analyzed. Aberrant promoter methylation was detected in both preinvasive and invasive lesions for genes APC, CDH1, CTNNB1, TIMP3, ESR1, and GSTP1. However, hierarchical mixed model and Generalized Estimating Equations model analyses showed that only APC, CDH1, and CTNNB1 promoter regions showed a higher frequency and methylation levels in pathologic samples when compared with normal breast. Whereas APC and CTNNB1 did not show differences in methylation levels or frequencies, CDH1 showed higher methylation levels in invasive tumors as compared with preinvasive lesions (P < 0.04, Mann-Whitney test with permutation correction). The analysis of APC, CDH1, and CTNNB1 methylation status was able to distinguish between normal and pathologic samples with a sensitivity of 67% (95% confidence interval, 60-71%) and a specificity of 75% (95% confidence interval, 69-81%). Our data point to the direct involvement of APC, CDH1, and CTNNB1 promoter methylation in the early stages of breast cancer progression and suggest that they may represent a useful tool for the detection of tumor cells in clinical specimens. (Cancer Epidemiol Biomarkers Prev 2009;18(10):2694–700)


Epigenetics | 2011

Regulation of KEAP1 expression by promoter methylation in malignant gliomas and association with patient’s outcome

Lucia Anna Muscarella; Raffaela Barbano; D'Angelo; Massimiliano Copetti; Michelina Coco; Balsamo T; la Torre A; Notarangelo A; Troiano M; Parisi S; Icolaro N; Catapano D; Vanna Maria Valori; Pellegrini F; Giuseppe Merla; Carella M; Vito Michele Fazio; Paola Parrella

In light with the view that KEAP1 loss of function may impact tumour behavior and modify response to chemotherapeutical agents, we sought to determine whether KEAP1 gene is epigenetically regulated in malignant gliomas. We developed a Quantitative Methylation Specific PCR (QMSP) assay to analyze 86 malignant gliomas and 20 normal brain tissues. The discriminatory power of the assay was assessed by Receiving Operating Characteristics (ROC) curve analysis. The AUC value of the curve was 0.823 (95%CI: 0.764-0.883) with an optimal cut off value of 0.133 yielding a 74% sensitivity (95%CI: 63%-82%) and an 85% specificity (95%CI: 64%-95%). Bisulfite sequencing analysis confirmed QMSP results and demonstrated a direct correlation between percentage of methylated CpGs and methylation levels (Spearman’s Rho 0.929, P=0.003). Remarkably, a strong inverse correlation was observed between methylation levels and KEAP1 mRNA transcript in tumour tissue (Spearman’s Rho -0.656 P=0.0001) and in a cell line before and after treatment with 5-azacytidine (P=0.003). RECPAM multivariate statistical analysis studying the interaction between MGMT and KEAP1 methylation in subjects treated with radiotherapy and temozolomide (n=70), identified three prognostic classes of glioma patients at different risk to progress. While simultaneous methylation of MGMT and KEAP1 promoters was associated with the lowest risk to progress, patients showing only MGMT methylation were the subgroup at the higher risk (HR 5.54, 95% CI 1.35-22.74). Our results further suggest that KEAP1 expression is epigenetically regulated. In addition we demonstrated that KEAP1 is frequently methylated in malignant gliomas and a predictor of patient’s outcome.


Epigenetics | 2013

Aberrant Keap1 methylation in breast cancer and association with clinicopathological features

Raffaela Barbano; Lucia Anna Muscarella; Barbara Pasculli; Vanna Maria Valori; Andrea Fontana; Michelina Coco; Annamaria la Torre; Teresa Balsamo; Maria Luana Poeta; Giovanni Francesco Marangi; Evaristo Maiello; Marina Castelvetere; Fabio Pellegrini; Roberto Murgo; Vito Michele Fazio; Paola Parrella

Keap1 (Kelch-like ECH-associated protein 1) is an adaptor protein that mediates the ubiquitination/degradation of genes regulating cell survival and apoptosis under oxidative stress conditions. We determined methylation status of the KEAP1 promoter in 102 primary breast cancers, 14 pre-invasive lesions, 38 paired normal breast tissues and 6 normal breast from reductive mammoplasty by quantitative methylation specific PCR (QMSP). Aberrant promoter methylation was detected in 52 out of the 102 primary breast cancer cases (51%) and 10 out of 14 pre-invasive lesions (71%). No mutations of the KEAP1 gene were identified in the 20 breast cancer cases analyzed by fluorescence based direct sequencing. Methylation was more frequent in the subgroup of patients identified as ER positive-HER2 negative tumors (66.7%) as compared with triple-negative breast cancers (35%) (p = 0.05, Chi-square test). The impact of the interactions between Er, PgR, Her2 expression and KEAP1 methylation on mortality was investigated by RECPAM multivariable statistical analysis, identifying four prognostic classes at different mortality risks. Triple-negative breast cancer patients with KEAP1 methylation had higher mortality risk than patients without triple-negative breast cancer (HR = 14.73, 95%CI: 3.65–59.37). Both univariable and multivariable COX regressions analyses showed that KEAP1 methylation was associated with a better progression free survival in patients treated with epirubicin/cyclophosfamide and docetaxel as sequential chemotherapy (HR = 0.082; 95%CI: 0.007–0.934). These results indicate that aberrant promoter methylation of the KEAP1 gene is involved in breast cancerogenesis. In addition, identifying patients with KEAP1 epigenetic abnormalities may contribute to disease progression prediction in breast cancer patients.


Virus Research | 2011

Hypermethylated levels of E-cadherin promoter in Huh-7 cells expressing the HCV core protein

Maria Ripoli; Raffaela Barbano; Teresa Balsamo; Claudia Piccoli; Virgilio Brunetti; Michelina Coco; Gianluigi Mazzoccoli; Manlio Vinciguerra; Valerio Pazienza

BACKGROUND AND AIM The mechanisms of hepatocarcinogenesis induced by hepatitis C virus remain unclear. Our aim was to investigate the effect of the HCV core protein on the promoter methylation status of selected genes potentially involved in the hepatocellular carcinoma (HCC). MATERIALS AND METHODS We evaluated the promoter methylation levels of the E-cadherin (CDH1), the glutathione S-transferase p1 (GSTP1), adenomatosis polyposis coli (APC), tissue inhibitor of metalloproteinase 3 (TIMP3), catenin (cadherin-associated protein) beta 1 (CNNTB1) genes by a quantitative methylation-specific polymerase chain reaction (QMSP) in the in vitro model of Huh-7 cells expressing the HCV core protein of genotype 1b. RESULTS We found that CDH1 promoter was hypermethylated in genotype 1b HCV core protein-positive cells as compared to control cells expressing the GFP protein alone (HCV core 1b vs GFP p=0.00; HCV core 1b vs Huh-7 p=0.03). This resulted in reduced levels of CDH1 protein as evaluated by immunoblot and by immunofluorescence. On the other hand no significant changes were observed for the other genes investigated. Furthermore, we present evidence that genotype 1b HCV core protein expression induces SIRT1 upregulation and that treatment with SIRT1 inhibitor sirtinol decreases the methylation levels of CDH1 promoter (1b+sirtinol vs 1b p=0.05; 1b+sirtinol vs GFP+sirtinol p=NS) resulting in 1.7-fold increased CDH1 mRNA expression (1b+sirtinol vs 1b p=0.05). CONCLUSIONS Our findings suggest that HCV core protein could play a role in HCC at least in part by altering the methylation status of CDH1 promoter. These findings could also suggest a novel therapeutic approach for HCC.


BioMed Research International | 2009

High Specificity of Quantitative Methylation-Specific PCR Analysis for MGMT Promoter Hypermethylation Detection in Gliomas

Paola Parrella; Antonella la Torre; Massimiliano Copetti; Vanna Maria Valori; Raffaela Barbano; Angelo Notarangelo; Michele Bisceglia; A. Gallo; Teresa Balsamo; Maria Luana Poeta; Massimo Carella; Domenico Catapano; Salvatore Parisi; Bruno Dallapiccola; Evaristo Maiello; Vincenzo D'Angelo; Vito Michele Fazio

Normal brain tissue from 28 individuals and 50 glioma samples were analyzed by real-time Quantitative Methylation-Specific PCR (QMSP). Data from this analysis were compared with results obtained on the same samples by MSP. QMSP analysis demonstrated a statistically significant difference in both methylation level (P = .000009 Mann Whitney Test) and frequencies (P = .0000007, Z-test) in tumour samples as compared with normal brain tissues. Although QMSP and MSP showed similar sensitivity, the specificity of QMSP analysis was significantly higher (93%; CI95%: 84%–100%) as compared with MSP (64%; 95%CI: 46%–82%). Our results suggest that QMSP analysis may represent a powerful tool to identify glioma patients that will benefit from alkylating agents chemotherapy.


Endocrine | 2012

A rare S33C mutation of CTNNB1 encoding β-catenin in a parathyroid adenoma found in an Italian primary hyperparathyroid cohort

Vito Guarnieri; Filomena Baorda; Claudia Battista; Michele Bisceglia; Teresa Balsamo; Elisa Gruppioni; Michelangelo Fiorentino; Lucia Anna Muscarella; Michelina Coco; Raffaela Barbano; Sabrina Corbetta; Anna Spada; David E. C. Cole; Lucie Canaff; Geoffrey N. Hendy; Massimo Carella; Alfredo Scillitani

Primary hyperparathyroidism (PHPT) is one of the most common endocrine disorders with a prevalence of 21/1000 in women between 55 and 75 years of age, corresponding to a 3/1000 prevalence in the general population [1]. Sporadic (non-familial) PHPT accounts for 90–95% of all cases. While activation of the protooncogene cyclin D1 (CCDN1) and inactivation of the tumor suppressor menin gene (MEN1) contribute to parathyroid adenomatosis, and inactivation of the parafibromin gene (CDC73) contributes to parathyroid carcinogenesis, the molecular pathogenesis of these tumors is incompletely understood. Dysregulated Wnt signaling and activation of b-catenin is involved in several cancers and consequently there has been recent interest in whether this is implicated in parathyroid tumorigenesis. In the absence of Wnt, cytosolic b-catenin phosphorylation catalyzed by glycogen synthase kinase (GSK)-b on serine/threonine residues (S33, S37, T41) encoded by exon 3 of the CTNNB1 gene leads to the ubiquitination and to degradation mediated by the proteasome. Mutation of the serine/threonine residues causes stabilization of the b-catenin protein and localization to the nucleus where it activates gene transcription. Several studies have been reported examining CTNNB1 gene mutations and aberrant b-catenin localization in sporadic parathyroid adenomas. While studies from Sweden found the homozygous S37A mutation in *7% of 124 parathyroid adenomas and aberrant b-catenin localization in all cases [2, 3], other studies from Japan [4, 5], USA [6],


Analytical Cellular Pathology | 2009

Comparison between real-time quantitative PCR detection of HER2 mRNA copy number in peripheral blood and ELISA of serum HER2 protein for determining HER2 status in breast cancer patients

Maria Savino; Paola Parrella; Massimiliano Copetti; Raffaela Barbano; Roberto Murgo; Vito Michele Fazio; Vanna Maria Valori; Massimo Carella; Maria Garrubba; Stefano Angelo Santini

Background: The development of non-invasive procedure to determine HER2 status may represent a powerful method for monitoring disease progression and response to the treatment. Methods: Serum samples and RNA from peripheral blood were evaluated in 85 breast cancer patients (49 HER2 positive and 36 HER2 negative) and 22 healthy controls. HER2 mRNA levels were measured by real-time quantitative PCR (QPCR) and serum HER2 protein by immunoenzimatic assay (EIA). ROC curve analyses were used to determine the optimal cut off values. Results: A statistically significant difference was detected for both QPCR and EIA in HER2 positive patients as compared with both healthy controls and HER2 negative tumours. QPCR showed a 91% (CI95%: 84%–98%) specificity and a 78% (CI95%: 68%–88%) sensitivity for an optimal cut off value of 4.74. The optimal cut off value for EIA was 22 ng/ml yielding a 95% (CI95%: 90%–100%) specificity and a 59% (CI95%: 48%–70%) sensitivity. The QPCR assay was slightly less specific than EIA in discriminating HER2 positive breast cancers from HER2 negative tumours (78% CI95%: 69%–87% versus 86% CI95%: 79%–93%), but it was more sensitive (76% CI95%: 67%–85% versus 55% CI95%: 44%–66%). Conclusions: Our results indicate that QPCR performs better than EIA in the determination of HER2 status of breast cancer patients and could be useful in monitoring the disease during follow up.


BMC Cancer | 2015

TRIM8 downregulation in glioma affects cell proliferation and it is associated with patients survival.

Lucia Micale; Carmela Fusco; Andrea Fontana; Raffaela Barbano; Bartolomeo Augello; Pasquelena De Nittis; Massimiliano Copetti; Maria Teresa Pellico; Barbara Mandriani; Dario Cocciadiferro; Paola Parrella; Vito Michele Fazio; Lucia Dimitri; Vincenzo D’Angelo; Chiara Novielli; Lidia Larizza; Antonio Daga; Giuseppe Merla

BackgroundHuman gliomas are a heterogeneous group of primary malignant brain tumors whose molecular pathogenesis is not yet solved. In this regard, a major research effort has been directed at identifying novel specific glioma-associated genes. Here, we investigated the effect of TRIM8 gene in glioma.MethodsTRIM8 transcriptional level was profiled in our own glioma cases collection by qPCR and confirmed in the independent TCGA glioma cohort. The association between TRIM8 expression and Overall Survival and Progression-free Survival in TCGA cohort was determined by using uni-multivariable Cox regression analysis. The effect of TRIM8 on patient glioma cell proliferation was evaluated by performing MTT and clonogenic assays. The mechanisms causing the reduction of TRIM8 expression were explored by using qPCR and in vitro assays.ResultsWe showed that TRIM8 expression correlates with unfavorable clinical outcome in glioma patients. We found that a restored TRIM8 expression induced a significant reduction of clonogenic potential in U87MG and patient’s glioblastoma cells. Finally we provide experimental evidences showing that miR-17 directly targets the 3′ UTR of TRIM8 and post-transcriptionally represses the expression of TRIM8.ConclusionsOur study provides evidences that TRIM8 may participate in the carcinogenesis and progression of glioma and that the transcriptional repression of TRIM8 might have potential value for predicting poor prognosis in glioma patients.


Seminars in Cancer Biology | 2018

Epigenetics of breast cancer: Biology and clinical implication in the era of precision medicine

Barbara Pasculli; Raffaela Barbano; Paola Parrella

In the last years, mortality from breast cancer has declined in western countries as a consequence of a more widespread screening resulting in earlier detection, as well as an improved molecular classification and advances in adjuvant treatment. Nevertheless, approximately one third of breast cancer patients will develop distant metastases and eventually die for the disease. There is now a compelling body of evidence suggesting that epigenetic modifications comprising DNA methylation and chromatin remodeling play a pivotal role since the early stages of breast cancerogenesis. In addition, recently, increasing emphasis is being placed on the property of ncRNAs to finely control gene expression at multiple levels by interacting with a wide array of molecules such that they might be designated as epigenetic modifiers. In this review, we summarize the current knowledge about the involvement of epigenetic modifications in breast cancer, and provide an overview of the significant association of epigenetic traits with the breast cancer clinicopathological features, emphasizing the potentiality of epigenetic marks to become biomarkers in the context of precision medicine.

Collaboration


Dive into the Raffaela Barbano's collaboration.

Top Co-Authors

Avatar

Paola Parrella

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Vito Michele Fazio

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Vanna Maria Valori

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Evaristo Maiello

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Massimiliano Copetti

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Barbara Pasculli

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Michelina Coco

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Lucia Anna Muscarella

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Teresa Balsamo

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar

Roberto Murgo

Casa Sollievo della Sofferenza

View shared research outputs
Researchain Logo
Decentralizing Knowledge