Network


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

Hotspot


Dive into the research topics where Sabrina Battista is active.

Publication


Featured researches published by Sabrina Battista.


Oncogene | 1997

Expression of the neoplastic phenotype by human thyroid carcinoma cell lines requires NFkappaB p65 protein expression.

Roberta Visconti; Janete Cerutti; Sabrina Battista; Monica Fedele; Francesco Trapasso; Kazuya Zeki; Maria Pia Miano; Filomena de Nigris; Laura Casalino; Francesco Curcio; Massimo Santoro; Alfredo Fusco

We have investigated the role of the NFκB complex in the process of thyroid carcinogenesis by analysing thyroid carcinoma cell lines. A significant increase in p65 NFκB mRNA and protein expression, compared to normal thyroid cultures or tissue, was found in all of the cancer cell lines. Conversely, only a modest increase in the p50 NFκB mRNA and protein was found in most, but not all carcinoma cell lines. The block of p65 protein synthesis with specific antisense oligonucleotides greatly reduced the ability of two undifferentiated carcinoma cell lines to form colonies in agar and reduced their growth rate. On the other hand, no effect was observed in the same cell lines when treated with p50 specific antisense oligonucleotides. These inhibitory effects seem to be mediated by the suppression of c-myc gene expression, since treatment with antisense oligonucleotides for p65 gene interfered negatively with c-myc gene expression. Our results indicate that activation of the NFκB complex by overexpression of p65 plays a critical role in the process of thyroid cell transformation.


Oncogene | 2002

Overexpression of the HMGA2 gene in transgenic mice leads to the onset of pituitary adenomas

Monica Fedele; Sabrina Battista; Lawrence Kenyon; Gustavo Baldassarre; Vincenzo Fidanza; Andres J. Klein-Szanto; A F Parlow; Rosa Visone; Giovanna Maria Pierantoni; Eric Outwater; Massimo Santoro; Carlo M. Croce; Alfredo Fusco

Overexpression of the HMGA2 gene is a common feature of neoplastic cells both in experimental and human models. Intragenic and extragenic HMGA2 rearrangements responsible for HMGA2 gene overexpression have been frequently detected in human benign tumours of mesenchymal origin. To better understand the role of HMGA2 overexpression in human tumorigenesis, we have generated transgenic mice carrying the HMGA2 gene under the transcriptional control of the cytomegalovirus promoter. High expression of the transgene was demonstrated in all the mouse tissues analysed, whereas no expression of the endogenous HMGA2 gene was detected in the same tissues from wild-type mice. In this study, two indipendent lines of transgenic mice have been generated. By 6 months of age, 85% of female animals of both transgenic lines developed pituitary adenomas secreting prolactin and growth hormone. The transgenic males developed the same phenotype with a lower penetrance (40%) and a longer latency period (about 18 months). Therefore, these data demonstrate that the overexpression of HMGA2 leads to the onset of mixed growth hormone/prolactin cell pituitary adenomas. These transgenic mice may represent an important tool for the study of this kind of neoplasia.


The EMBO Journal | 1997

Neoplastic transformation of rat thyroid cells requires the junB and fra-1 gene induction which is dependent on the HMGI-C gene product.

Daniela Vallone; Sabrina Battista; Giovanna Maria Pierantoni; Monica Fedele; Laura Casalino; Massimo Santoro; Giuseppe Viglietto; Alfredo Fusco; Pasquale Verde

The expression of the high mobility group I (HMGI)‐C chromatin component was shown previously to be essential for the establishment of the neoplastic phenotype in retrovirally transformed thyroid cell lines. To identify possible targets of the HMGI‐C gene product, we have analyzed the AP‐1 complex in normal, fully transformed and antisense HMGI‐C‐expressing rat thyroid cells. We show that neoplastic transformation is associated with a drastic increase in AP‐1 activity, which reflects multiple compositional changes. The strongest effect is represented by the dramatic junB and fra‐1 gene induction, which is prevented in cell lines expressing the antisense HMGI‐C. These results indicate that the HMGI‐C gene product is essential for the junB and fra‐1 transcriptional induction associated with neoplastic transformation. The inhibition of Fra‐1 protein synthesis by stable transfection with a fra‐1 antisense RNA vector significantly reduces the malignant phenotype of the transformed thyroid cells, indicating a pivotal role for the fra‐1 gene product in the process of cellular transformation.


Oncogene | 2005

Transgenic mice overexpressing the wild-type form of the HMGA1 gene develop mixed growth hormone/prolactin cell pituitary adenomas and natural killer cell lymphomas

Monica Fedele; Francesca Pentimalli; Gustavo Baldassarre; Sabrina Battista; Andres J. Klein-Szanto; Lawrence Kenyon; Rosa Visone; Ivana De Martino; Andrea Ciarmiello; Claudio Arra; Giuseppe Viglietto; Carlo M. Croce; Alfredo Fusco

Overexpression of HMGA1 proteins is a constant feature of human carcinomas. Moreover, rearrangements of this gene have been detected in several human benign tumors of mesenchymal origin. To define the role of these proteins in cell transformation in vivo, we have generated transgenic mice overexpressing ubiquitously the HMGA1 gene. These mice developed mixed growth hormone/prolactin cell pituitary adenomas and natural killer (NK)-T/NK cell lymphomas. The HMGA1-induced expression of IL-2 and IL-15 proteins and their receptors may account for the onset of these lymphomas. At odds with mice overexpressing a wild-type or a truncated HMGA2 protein, adrenal medullar hyperplasia and pancreatic islet cell hyperplasia frequently occurred and no increase in body size and weight was observed in HMGA1 mice. Taken together, these data indicate an oncogenic role of the HMGA1 gene also in vivo.


Molecular and Cellular Biology | 2003

Negative Regulation of BRCA1 Gene Expression by HMGA1 Proteins Accounts for the Reduced BRCA1 Protein Levels in Sporadic Breast Carcinoma

Gustavo Baldassarre; Sabrina Battista; Barbara Belletti; Sanjay Thakur; Francesca Pentimalli; Francesco Trapasso; Monica Fedele; Giovanna Maria Pierantoni; Carlo M. Croce; Alfredo Fusco

ABSTRACT A drastic reduction in BRCA1 gene expression is a characteristic feature of aggressive sporadic breast carcinoma. However, the mechanisms underlying BRCA1 downregulation in breast cancer are not well understood. Here we report that both in vitro and in vivo HMGA1b protein binds to and inhibits the activity of both human and mouse BRCA1 promoters. Consistently, murine embryonic stem (ES) cells with the Hmga1 gene deleted display higher Brca1 mRNA and protein levels than do wild-type ES cells. Stable transfection of MCF-7 cells with the HMGA1b cDNA results in a decrease of BRCA1 gene expression and in a lack of BRCA1 induction after estrogen treatment. Finally, we found an inverse correlation between HMGA1 and BRCA1 mRNA and protein expression in human mammary carcinoma cell lines and tissues. These data indicate that HMGA1 proteins are involved in transcriptional regulation of the BRCA1 gene, and their overexpression may have a role in BRCA1 downregulation observed in aggressive mammary carcinomas.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Onset of natural killer cell lymphomas in transgenic mice carrying a truncated HMGI-C gene by the chronic stimulation of the IL-2 and IL-15 pathway

Gustavo Baldassarre; Monica Fedele; Sabrina Battista; Andrea Vecchione; Andres J. Klein-Szanto; Massimo Santoro; Thomas A. Waldmann; Nazli Azimi; Carlo M. Croce; Alfredo Fusco

Rearrangements of the high mobility group protein I-C (HMGI-C) gene, consisting in the loss of the carboxyl-terminal tail, have been frequently detected in benign human tumors of mesenchymal origin. We have previously demonstrated that transgenic (TG) mice carrying a truncated HMGI-C construct (HMGI-C/T) exhibit a giant phenotype together with a predominantly abdominal/pelvic lipomatosis. Here, we report that HMGI-C/T TG mice develop natural killer (NK)-T/NK cell lymphomas starting from 12 months of age. We found an increased expression of IL-2 and IL-15 proteins and their receptors in these lymphomas, and we demonstrate that HMGI-C/T protein positively regulates their expression in vitro. Therefore, the HMGI-C/T-mediated chronic stimulation of the IL-2/IL-15 pathway could be responsible for the onset of NK-T/NK cell lymphomas in HMGI-C/T TG mice.


Molecular and Cellular Biology | 2001

Critical role of the HMGI(Y) proteins in adipocytic cell growth and differentiation.

Rosa Marina Melillo; Giovanna Maria Pierantoni; Stefania Scala; Sabrina Battista; Monica Fedele; Antonella Stella; M. C. De Biasio; Gennaro Chiappetta; V. Fidanza; Gianluigi Condorelli; Massimo Santoro; C. M. Croce; Giuseppe Viglietto; Alfredo Fusco

ABSTRACT The high-mobility group I (HMGI) nonhistone chromosomal proteins HMGI(Y) and HMGI-C have been implicated in defining chromatin structure and in regulating the transcription of several genes. These proteins have been implicated in adipocyte homeostasis: a severe deficiency of fat tissue is found in mice with targeted disruption of the HMGI-C locus, and lipomagenesis in humans is frequently associated with somatic mutations of HMGI genes. The aim of this study was to examine the role of HMGI(Y) proteins in adipocytic cell growth and differentiation. First, we found that differentiation of the preadipocytic 3T3-L1 cell line caused early induction of HMGI(Y) gene expression. Suppression of HMGI(Y) expression by antisense technology dramatically increased the growth rate and impaired adipocytic differentiation in these cells. The process of adipogenic differentiation involves the interplay of several transcription factors, among which is the CCAAT/enhancer-binding protein (C/EBP) family of proteins. These factors are required for the transcriptional activation of adipocyte-specific genes. We also tested the hypothesis that HMGI(Y) might participate in transcriptional control of adipocyte-specific promoters. We found that HMGI(Y) proteins bind C/EBPβ in vivo and in vitro. Furthermore, we show that HMGI(Y) strongly potentiates the capacity of C/EBPβ to transactivate the leptin promoter, an adipose-specific promoter. Taken together, these results indicate that the HMGI(Y) proteins play a critical role in adipocytic cell growth and differentiation.


Cancer Research | 2006

Haploinsufficiency of the Hmga1 gene causes cardiac hypertrophy and myelo-lymphoproliferative disorders in mice.

Monica Fedele; Vincenzo Fidanza; Sabrina Battista; Francesca Pentimalli; Andres J. Klein-Szanto; Rosa Visone; Ivana De Martino; Antonio Curcio; Carmine Morisco; Luigi Del Vecchio; Gustavo Baldassarre; Claudio Arra; Giuseppe Viglietto; Ciro Indolfi; Carlo M. Croce; Alfredo Fusco

The HMGA1 protein is a major factor in chromatin architecture and gene control. It plays a critical role in neoplastic transformation. In fact, blockage of HMGA1 synthesis prevents rat thyroid cell transformation by murine transforming retroviruses, and an adenovirus carrying the HMGA1 gene in the antisense orientation induces apoptotic cell death in anaplastic human thyroid carcinoma cell lines, but not in normal thyroid cells. Moreover, both in vitro and in vivo studies have established the oncogenic role of the HMGA1 gene. In this study, to define HMGA1 function in vivo, we examined the consequences of disrupting the Hmga1 gene in mice. Both heterozygous and homozygous mice for the Hmga1-null allele show cardiac hypertrophy due to the direct role of HMGA1 on cardiomyocytic cell growth regulation. These mice also developed hematologic malignancies, including B cell lymphoma and myeloid granuloerythroblastic leukemia. The B cell expansion and the increased expression of the RAG1/2 endonuclease, observed in HMGA1-knockout spleen tissues, might be responsible for the high rate of abnormal IgH rearrangements observed in these neoplasias. Therefore, the data reported here indicate the critical role of HMGA1 in heart development and growth, and reveal an unsuspected antioncogenic potential for this gene in hematologic malignancies.


Oncogene | 2002

HMGA1 and HMGA2 protein expression in mouse spermatogenesis.

Paolo Chieffi; Sabrina Battista; Marco Barchi; Silvia Di Agostino; Giovanna Maria Pierantoni; Monica Fedele; Lorenzo Chiariotti; Donatella Tramontano; Alfredo Fusco

The high-mobility group A (HMGA) nonhistone chromosomal proteins HMGA1 and HMGA2 play a role in determining chromatin structure and in regulating the transcription of several genes. High levels of these proteins are characteristic of rapidly dividing cells in embryonic tissue and in tumors. The aim of this study was to determine the role of HMGA1 and HMGA2 throughout mouse spermatogenesis. Northern blot analysis and immunocytochemistry showed HMGA1 and HMGA2 expression during the progression from spermatocyte to spermatid. Interestingly, Western blot analysis with antibodies against the HMGA1 gene product revealed only the HMG1c isoform (27 kDa) in the testis; HMGA1a and HMGA1b were undetectable. These three isoforms are encoded by the HMGA1 gene through alternative splicing. Finally, few spermatids and complete absence of spermatozoa were observed in the testes of HMGA2-null mice, which suggests that the HMGA2 gene plays a critical role in male fertility.


Nature Reviews Endocrinology | 2014

Deregulation of microRNA expression in thyroid neoplasias

Pierlorenzo Pallante; Sabrina Battista; Giovanna Maria Pierantoni; Alfredo Fusco

MicroRNAs (miRNAs) have emerged as a class of powerful gene expression regulators. Acting at the post-transcriptional level, miRNAs modulate the expression of at least one-third of the mRNAs that are encoded by the human genome. The expression of a single gene can be regulated by several miRNAs, and every miRNA has more than one target gene. Thus, the miRNA regulatory circuit, which affects essential cellular functions, is of enormous complexity. Moreover, a fundamental role for miRNAs has been determined in the onset and progression of human cancers. Here, we summarize the main alterations in miRNA expression that have been identified in thyroid neoplasias and examine the mechanisms through which miRNA deregulation might promote thyroid cell transformation. We also discuss how the emerging knowledge on miRNA deregulation could be harnessed for the diagnosis and treatment of thyroid neoplasias.

Collaboration


Dive into the Sabrina Battista's collaboration.

Top Co-Authors

Avatar

Alfredo Fusco

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Monica Fedele

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giovanna Maria Pierantoni

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Gennaro Chiappetta

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gustavo Baldassarre

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marianna Colamaio

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Antonella Federico

University of Naples Federico II

View shared research outputs
Researchain Logo
Decentralizing Knowledge