Network


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

Hotspot


Dive into the research topics where Teresa Balsamo is active.

Publication


Featured researches published by Teresa Balsamo.


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.


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.


International Journal of Cancer | 2011

High RAD51 mRNA expression characterize estrogen receptor‐positive/progesteron receptor‐negative breast cancer and is associated with patient's outcome

Raffaela Barbano; Massimiliano Copetti; Giuseppe Perrone; Valerio Pazienza; Lucia Anna Muscarella; Teresa Balsamo; Clelia Tiziana Storlazzi; Maria Ripoli; Monica Rinaldi; Vanna Maria Valori; Tiziana Latiano; Evaristo Maiello; Pietro Stanziale; Massimo Carella; Alessandra Mangia; Fabio Pellegrini; Michele Bisceglia; Andrea Onetti Muda; Vittorio Altomare; Roberto Murgo; Vito Michele Fazio; Paola Parrella

Mutations in DNA double‐strand breaks (DSB) repair genes are involved in the pathogenesis of hereditary mammary tumors, it is, however, still unclear whether defects in this pathway may play a role in sporadic breast cancer. In this study, we initially determined mRNA expression of 15 DSB related genes by reverse transcription quantitative polymerase chain reaction in paired normal tissue and cancer specimen from 20 breast cancer cases to classify them into homogeneous clusters. G22P1/ku70, ATR and RAD51 genes were differentially expressed in the three branches recognized by clustering analysis. In particular, a breast cancer subgroup characterized by high RAD51 mRNA levels and estrogen receptor (ER)‐positive/progesteron receptor (PR)‐negative phenotype was identified. This result was confirmed by the analysis of G22P1/ku70, ATR and RAD51 mRNA levels on paired normal and tumor specimens from an extended breast cancer cohort (n = 75). RAD51 mRNA levels were inversely associated with PR status (p = 0.02) and the highest levels were, indeed, detected in ER‐positive/PR‐negative tumors (p = 0.03). RAD51 immunostaining of a tissue microarray confirmed the inverse relationship between high RAD51 expression and negative PR status (p = 0.002), as well as, the association with ER‐positive/PR‐negative phenotype (p = 0.003). Interestingly, the analysis of microarray expression data from 295 breast cancers indicate that RAD51 increased mRNA expression is associated with higher risk of tumor relapse, distant metastases and worst overall survival (p = 0.015, p = 0.009 and p = 0.013 respectively). Our results suggest that RAD51 expression determination could contribute to a better molecular classification of mammary tumors and may represent a novel tool for evaluating postoperative adjuvant therapy for breast cancer patients.


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.


Oncotarget | 2017

Keap1/Nrf2 pathway in kidney cancer: frequent methylation of KEAP1 gene promoter in clear renal cell carcinoma

Federico Pio Fabrizio; Manuela Costantini; Massimiliano Copetti; Annamaria la Torre; Angelo Sparaneo; Andrea Fontana; Luana Poeta; Michele Gallucci; Steno Sentinelli; Paolo Graziano; Paola Parente; Vincenzo Pompeo; Laura De Salvo; Giuseppe Simone; Rocco Papalia; Francesco Picardo; Teresa Balsamo; Gerardo Flammia; Domenico Trombetta; Angela Pantalone; Klaas Kok; Ferronika Paranita; Lucia Anna Muscarella; Vito Michele Fazio

The Keap1/Nrf2 pathway is a master regulator of the cellular redox state through the induction of several antioxidant defence genes implicated in chemotherapeutic drugs resistance of tumor cells. An increasing body of evidence supports a key role for Keap1/Nrf2 pathway in kidney diseases and renal cell carcinoma (RCC), but data concerning the molecular basis and the clinical effect of its deregulation remain incomplete. Here we present a molecular profiling of the KEAP1 and NFE2L2 genes in five different Renal Cell Carcinoma histotypes by analysing 89 tumor/normal paired tissues (clear cell Renal Carcinoma, ccRCCs; Oncocytomas; Papillary Renal Cell Carcinoma Type 1, PRCC1; Papillary Renal Cell Carcinoma Type 2, PRCC2; and Chromophobe Cell Carcinoma). A tumor-specific DNA methylation of the KEAP1 gene promoter region was found as a specific feature of the ccRCC subtype (18/37, 48.6%) and a direct correlation with mRNA levels was confirmed by in vitro 5-azacytidine treatment. Analysis of an independent data set of 481 ccRCC and 265 PRCC tumors corroborates our results and multivariate analysis reveals a significant correlation among ccRCCs epigenetic KEAP1 silencing and staging, grading and overall survival. Our molecular results show for the the first time the epigenetic silencing of KEAP1 promoter as the leading mechanism for modulation of KEAP1 expression in ccRCCs and corroborate the driver role of Keap1/Nrf2 axis deregulation with potential new function as independent epigenetic prognostic marker in renal cell carcinoma.


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],


Oncotarget | 2017

Frequent NRG1 fusions in caucasian pulmonary mucinous adenocarcinoma predicted by Phospho-ErbB3 expression

Domenico Trombetta; Paolo Graziano; Aldo Scarpa; Angelo Sparaneo; Giulio Rossi; Antonio Rossi; Massimo Di Maio; Davide Antonello; Andrea Mafficini; Federico Pio Fabrizio; Maria Carmina Manzorra; Teresa Balsamo; Flavia Centra; Michele Simbolo; Angela Pantalone; Michela Notarangelo; Paola Parente; Maria Cecilia Lucia Dimitri; Antonio Bonfitto; Fabiola Fiordelisi; Clelia Tiziana Storlazzi; Alberto L'Abbate; Marco Taurchini; Evaristo Maiello; Vito Michele Fazio; Lucia Anna Muscarella

NRG1 fusions were recently reported as a new molecular feature of Invasive Mucinous Adenocarcinoma (IMA) of the lung. The NRG1 chimeric ligand acts as a strong inductor of phosphorylation and tyrosine kinase activity of the ErbB2/ErbB3 heterodimer, thus enhancing the PI3K–AKT/MAPK pathways. The NRG1 fusions were widely investigated in Asian IMA cohorts, whereas just anecdotal information are available about the occurrence of NRG1 fusions in IMA Caucasian population. Here we firstly explored a large Caucasian cohort of 51 IMAs and 34 non-IMA cases for the occurrence of NRG1 rearrangements by fluorescent in situ hybridization (FISH) and RNA target sequencing. FISH results were correlated to the immunohistochemical expression of phosphorylated-ErbB3 (pErbB3) receptor and the mutational status of KRAS, EGFR and ALK genes. The NRG1 rearrangements were detected in 31% IMAs and 3% non-IMAs and the CD74-NRG1 fusion transcript variant was characterized in 4 NRG1-positive IMAs. Moreover, pErbB3 expression was found to be strictly associated to the mucinous pattern (p = 0.012, Chi-square test) and all IMA cases showing aberrant expression of pErbB3 demonstrated NRG1 rearrangements. No significant correlation between NRG1 rearrangements and EGFR, KRAS or ALK mutations respectively, was observed. We report for the first time that NRG1 fusions are driver alterations clearly associated with mucinous lung adenocarcinoma subtype of Caucasian patients and not exclusive of Asiatic population. pErbB3 immunostaining may represent a strong predictor of NRG1 fusions, pointing out the detection of pErbB3 by IHC as a rapid and effective pre-screening method to select the NRG1-positive patients.


Oncology Letters | 2017

Novel association of MEN1 gene mutations with parathyroid carcinoma

Luigia Cinque; Angelo Sparaneo; Filomena Cetani; Michelina Coco; Celeste Clemente; Massimiliano Chetta; Teresa Balsamo; Claudia Battista; Eliana Sanpaolo; Elena Pardi; Leonardo D'Agruma; Claudio Marcocci; Evaristo Maiello; Geoffrey N. Hendy; David E. C. Cole; Alfredo Scillitani; Vito Guarnieri

Inactivating mutations of the multiple endocrine neoplasia 1 (MEN1) gene cause MEN1 syndrome, characterized by primary hyperparathyroidism (pHPT), and parathyroid and gastro-entero-pancreatic pituitary tumors. At present, only 14 cases of malignant parathyroid tumor have been associated with the syndrome, with 6 cases carrying an inactivating mutation of the MEN1 gene. The present study presents the case of a 48-year-old female who presented with multigland pHPT and multiple pancreatic lesions. The patient underwent surgery several times for the excision of parathyroid hyperplasia, carcinoma and adenoma. The MEN1 gene was screened, revealing three variants (in cis) at the intron/exon 3 boundary (IVS2-3G>C, c.497A>T and c.499G>T) detected on the DNA of the proband, not shared by her relatives. RNA sequencing revealed that the IVS2-3C>G variant caused the skipping of the exon 3. Therefore, the present study reports on a novel rare association of MEN1 syndrome and parathyroid carcinoma. The reported splicing mutation was previously identified in subjects who always developed malignant lesions; thus, a possible genotype-phenotype association may be considered.


Cancer Research | 2014

Abstract 2251: nrf2-keap1 axis molecular profile in small cell lung cancer cell lines

Lucia Anna Muscarella; Annamaria la Torre; Angelo Sparaneo; Orazio Palumbo; Clelia Tiziana Storlazzi; Teresa Balsamo; Domenico Trombetta; Fabio Pellegrini; Raffaela Barbano; Barbara Pasculli; Paolo Graziano; Montse Sanchez-Cespedes; Maria Teresa Landi; Paola Parrella; Vito Michele Fazio

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The Keap1/Nrf2 pathway is a master regulator of antioxidants and cellular stress responses implicated in resistance of tumor cells against chemotherapeutic drugs. The link between molecular alterations of this pathway in Non Small Cell Lung Cancer (NSCLC) is well studied and appears to depend on several main factors including the existence of activating mutations in NFE2LE gene and/or loss of function mutations and methylation in the KEAP1 gene. At present, the data concerning the mechanism of alteration of Nrf2-Keap1 pathway in Small Cell Lung Cancer (SCLC) instead are almost incomplete and correlation analysis with therapeutic strategies targeting the molecular dysfunction of this pathway is ongoing. Here we present a comprehensive molecular alteration profile of the main partners of the Nrf2/keap1 axis in 12 SCLC cell lines by integrating data from SNP-Array analysis, immunofluorescence, mutation screening by direct sequencing, methylation by QMSP and expression analyses by RT-qPCR and western blotting. Our analyses confirm the global deregulation of Nrf2/Keap1 pathway in SCLC cell lines, showing an hypermethylation of the CpGs located into the P1 promoter region of the KEAP1 in 42% (5/12) of the cell lines, and a chromosomal amplification involving the NFE2LE gene locus (2q31.1) in two cell lines. Only one just described point mutation in the kelch-repeat 2 was observed in one of the cell line analysed. Our gene-alteration profile of SCLC cell lines provides new insights into the mechanism of deregulation of Nrf2-Keap1 detoxification pathway in this group of high grade neuroendocrine lung cancers, suggesting the NFE2LE gene amplification and the KEAP1 promoter hypermethylation as alternative mechanisms of dysfunctional Nrf2/Keap1 in SCLCs.Analyses on tumor tissues are ongoing to confirm these observations. Moreover, the provided full molecular data from cell lines will be useful for in vitro functional studies aimed to establish new combined therapeutic strategies in targeted cancer treatments of this aggressive lung tumor histotype. Citation Format: Lucia Anna Muscarella, Annamaria la Torre, Angelo Sparaneo, Orazio Palumbo, Clelia Tiziana Storlazzi, Teresa Balsamo, Domenico Trombetta, Fabio Pellegrini, Raffaela Barbano, Barbara Pasculli, Paolo Graziano, Montse Sanchez-cespedes, Maria Teresa Landi, Paola Parrella, Vito Michele Fazio. nrf2-keap1 axis molecular profile in small cell lung cancer cell lines. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2251. doi:10.1158/1538-7445.AM2014-2251

Collaboration


Dive into the Teresa Balsamo's collaboration.

Top Co-Authors

Avatar

Vito Michele Fazio

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

Raffaela Barbano

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

Paola Parrella

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

Vanna Maria Valori

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annamaria la Torre

Casa Sollievo della Sofferenza

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge