Network


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

Hotspot


Dive into the research topics where Francesca Carlomagno is active.

Publication


Featured researches published by Francesca Carlomagno.


Journal of Clinical Investigation | 1992

Ret oncogene activation in human thyroid neoplasms is restricted to the papillary cancer subtype.

Santoro M; Francesca Carlomagno; I D Hay; M A Herrmann; Michele Grieco; Rosa Marina Melillo; Marco A. Pierotti; Italia Bongarzone; G. Della Porta; N Berger

We have recently reported the activation of a new oncogene in human papillary thyroid carcinomas. This oncogene, papillary thyroid carcinoma (PTC), is a novel rearranged version of the ret tyrosine-kinase protooncogene. Thyroid neoplasms include a broad spectrum of malignant tumors, ranging from well-differentiated tumors to undifferentiated anaplastic carcinomas. To determine the frequency of ret oncogene activation, we analyzed 286 cases of human thyroid tumors of diverse histologic types. We found the presence of an activated form of the ret oncogene in 33 (19%) of 177 papillary carcinomas. By contrast, none of the other 109 thyroid tumors, which included 37 follicular, 15 anaplastic, and 18 medullary carcinomas, and 34 benign lesions, showed ret activation.


Oncogene | 2004

Disease associated mutations at valine 804 in the RET receptor tyrosine kinase confer resistance to selective kinase inhibitors.

Francesca Carlomagno; Teresa Guida; Suresh Anaganti; Giancarlo Vecchio; Alfredo Fusco; Anderson J. Ryan; Marc Billaud; Massimo Santoro

We have recently demonstrated that the pyrazolopyrimidines PP1 and PP2 and the 4-anilinoquinazoline ZD6474 display a strong inhibitory activity (IC50⩽100 nM) towards constitutively active oncogenic RET kinases. Here, we show that most oncogenic MEN2-associated RET kinase mutants are highly susceptible to PP1, PP2 and ZD6474 inhibition. In contrast, MEN2-associated swap of bulky hydrophobic leucine or methionine residues for valine 804 in the RET kinase domain causes resistance to the three compounds. Substitution of valine 804 with the small amino- acid glycine renders the RET kinase even more susceptible to inhibition (ZD6474 IC50: 20 nM) than the wild-type kinase. Our data identify valine 804 of RET as a structural determinant mediating resistance to pyrazolopyrimidines and 4-anilinoquinazolines.


Annals of the New York Academy of Sciences | 2006

Molecular Mechanisms of RET Activation in Human Cancer

Massimo Santoro; Rosa Marina Melillo; Francesca Carlomagno; Alfredo Fusco; Giancarlo Vecchio

Abstract: Mutations that produce oncogenes with dominant gain of function target receptor protein tyrosine kinases (PTKs) in cancer and confer uncontrolled proliferation, impaired differentiation, or unrestrained survival to the cancer cell. However, insufficient PTK signaling may be responsible for developmental diseases. Gain of function of the RET receptor PTK is associated with human cancer. At the germline level, point mutations of RET are responsible for multiple endocrine neoplasia type 2 (MEN2A, MEN2B, and FMTC). Mutations of extracellular cysteines are found in MEN2A patients, and a Met918Thr mutation is responsible for most MEN2B cases. At the somatic level, gene rearrangements juxtaposing the tyrosine kinase domain of RET to heterologous gene partners are found in papillary carcinomas of the thyroid. These rearrangements generate the chimeric RET/PTC oncogenes. Both MEN2 mutations and PTC gene rearrangements potentiate the intrinsic tyrosine kinase activity of RET and, ultimately, the RET downstream signaling events. A multidocking site of the C‐tail of RET is essential for both mitogenic and survival RET signaling. Such a site is involved in the recruitment of several intracellular molecules, such as the Shc, FRS2, IRS1, Gab1/2, and Enigma. The different activating mutations not only potentiate the enzymatic activity of the RET kinase but also may alter qualitatively RET signaling properties by: (1) altering RET autophosphorylation (in the case of the MEN2B mutation), (2) modifying the subcellular distribution of the active kinase, and (3) providing the active kinase with a scaffold for novel protein‐protein interactions (as in the case of RET/PTC oncoproteins). This review describes the molecular mechanisms by which the different genetic alterations cause the conversion of RET into a dominant transforming oncogene.


Oncogene | 1998

Signalling of the Ret receptor tyrosine kinase through the c-Jun NH2-terminal protein kinases (JNKS): evidence for a divergence of the ERKs and JNKs pathways induced by Ret.

Mario Chiariello; Roberta Visconti; Francesca Carlomagno; Rosa Marina Melillo; Cecilia Bucci; Vittorio de Franciscis; Gary M Fox; Shuqian Jing; Omar A. Coso; J. Silvio Gutkind; Alfredo Fusco; Massimo Santoro

The RET proto-oncogene encodes a functional receptor tyrosine kinase (Ret) for the Glial cell line Derived Neurotrophic Factor (GDNF). RET is involved in several neoplastic and non-neoplastic human diseases. Oncogenic activation of RET is detected in human papillary thyroid tumours and in multiple endocrine neoplasia type 2 syndromes. Inactivating mutations of RET have been associated to the congenital megacolon, i.e. Hirschprungs disease. In order to identify pathways that are relevant for Ret signalling to the nucleus, we have investigated its ability to induce the c-Jun NH2-terminal protein kinases (JNK). Here we show that triggering the endogenous Ret, expressed in PC12 cells, induces JNK activity; moreover, Ret is able to activate JNK either when transiently transfected in COS-1 cells or when stably expressed in NIH3T3 fibroblasts or in PC Cl 3 epithelial thyroid cells. JNK activation is dependent on the Ret kinase function, as a kinase-deficient RET mutant, associated with Hirschsprungs disease, fails to activate JNK. The pathway leading to the activation of JNK by RET is clearly divergent from that leading to the activation of ERK: substitution of the tyrosine 1062 of Ret, the Shc binding site, for phenylalanine abrogates ERK but not JNK activation. Experiments conducted with dominant negative mutants or with negative regulators demonstrate that JNK activation by Ret is mediated by Rho/Rac related small GTPases and, particularly, by Cdc42.


The EMBO Journal | 1996

Molecular heterogeneity of RET loss of function in Hirschsprung's disease.

Francesca Carlomagno; G De Vita; Maria Teresa Berlingieri; V. De Franciscis; Rosa Marina Melillo; V. Colantuoni; Matthias H. Kraus; P P Di Fiore; A Fusco; Massimo Santoro

The RET proto‐oncogene encodes a receptor with tyrosine kinase activity (RET) that is involved in several neoplastic and non‐neoplastic diseases. Oncogenic activation of RET, achieved by different mechanisms, is detected in a sizeable fraction of human thyroid tumors, as well as in multiple endocrine neoplasia types 2A and 2B (MEN2A and MEN2B) and familial medullary thyroid carcinoma tumoral syndromes. Germline mutations of RET have also been associated with a non‐neoplastic disease, the congenital colonic aganglionosis, i.e. Hirschsprungs disease (HSCR). To analyse the impact of HSCR mutations on RET function, we have introduced into wild‐type RET and activated RET(MEN2A) and RET(MEN2B) alleles three missense mutations associated with HSCR. Here we show that the three mutations caused a loss of function of RET when assayed in two model cell systems, NIH 3T3 and PC12 cells. The effect of different HSCR mutations was due to different molecular mechanisms. The HSCR972 (Arg972–>Gly) mutation, mapping in the intracytoplasmic region of RET, impaired its tyrosine kinase activity, while two extracellular mutations, HSCR32 (Ser32–>Leu) and HSCR393 (Phe393–>Leu), inhibited the biological activity of RET by impairing the correct maturation of the RET protein and its transport to the cell surface.


Oncogene | 2004

Functional expression of the CXCR4 chemokine receptor is induced by RET/PTC oncogenes and is a common event in human papillary thyroid carcinomas.

Maria Domenica Castellone; Valentina Guarino; Valentina De Falco; Francesca Carlomagno; Fulvio Basolo; Pinuccia Faviana; Mogens Kruhøffer; Torben F. Ørntoft; John P Russell; Jay L. Rothstein; Alfredo Fusco; Massimo Santoro; Rosa Marina Melillo

To identify genes involved in the transformation of thyroid follicular cells, we explored, using DNA oligonucleotide microarrays, the transcriptional response of PC Cl3 rat thyroid epithelial cells to the ectopic expression of the RET/PTC oncogenes. We found that RET/PTC was able to induce the expression of CXCR4, the receptor for the chemokine CXCL12/SDF-1α/β. We observed that CXCR4 expression correlated with the transforming ability of the oncoprotein and depended on the integrity of the RET/PTC–RAS/ERK signaling pathway. We found that CXCR4 was expressed in RET/PTC-positive human thyroid cancer cell lines, but not in normal thyroid cells. Furthermore, we found CXCR4 expression in human thyroid carcinomas, but not in normal thyroid samples by immunohistochemistry. Since CXCR4 has been recently implicated in tumor proliferation, motility and invasiveness, we asked whether treatment with SDF-1α was able to induce a biological response in thyroid cells. We observed that SDF-1α induced S-phase entry and survival of thyroid cells. Invasion through a reconstituted extracellular matrix was also supported by SDF-1α and inhibited by a blocking antibody to CXCR4. Taken together, these results suggest that human thyroid cancers bearing RET/PTC rearrangements may use the CXCR4/SDF-1α receptor–ligand pathway to proliferate, survive and migrate.


Oncogene | 2002

Cloning and molecular characterization of a novel gene strongly induced by the adenovirus E1A gene in rat thyroid cells.

Roberta Visconti; Filippo Schepis; Rodolfo Iuliano; Giovanna Maria Pierantoni; Li Zhang; Francesca Carlomagno; Caterina Battaglia; Maria Luisa Martelli; Francesco Trapasso; Massimo Santoro; Alfredo Fusco

Expression of the adenovirus E1A gene in the rat thyroid differentiated cell line PC Cl 3 induces thyrotropin-independent cell growth and impairs differentiation. However, the malignant phenotype is achieved only when the PC E1A cells are infected with other murine retroviruses carrying the v-abl, v-raf or polyoma middle-T genes. To determine through which genes E1A affects thyroid cells, we differentially screened PC Cl 3 and PC E1A cells. Here we report a new gene, named CL2, that is upregulated in PC E1A cells. The CL2 transcript is 4.4 kb long and encodes a 949 amino-acid protein. Conceptual translation of the open reading frame showed one product with a signal peptide, multiple nuclear localization signals and three newly described domains. Furthermore, in vivo, this protein was located juxtanuclear, which is suggestive of Golgian localization, and also in cytoplasm and nucleus/nucleolus. Finally, CL2 gene expression was drastically downregulated in human thyroid neoplastic cell lines and tissues.


American Journal of Pathology | 2002

Potent Mitogenicity of the RET/PTC3 Oncogene Correlates with Its Prevalence in Tall-Cell Variant of Papillary Thyroid Carcinoma

Fulvio Basolo; Riccardo Giannini; Carmen Monaco; Rosa Marina Melillo; Francesca Carlomagno; Martina Pancrazi; Giuliana Salvatore; Gennaro Chiappetta; Furio Pacini; Rossella Elisei; Paolo Miccoli; Aldo Pinchera; Alfredo Fusco; Massimo Santoro

The tall-cell variant (TCV) of papillary thyroid carcinoma (PTC), characterized by tall cells bearing an oxyphilic cytoplasm, is more clinically aggressive than conventional PTC. RET tyrosine kinase rearrangements, which represent the most frequent genetic alteration in PTC, lead to the recombination of RET with heterologous genes to generate chimeric RET/PTC oncogenes. RET/PTC1 and RET/PTC3 are the most prevalent variants. We have found RET rearrangements in 35.8% of TCV (14 of 39 cases). Whereas the prevalences of RET/PTC1 and RET/PTC3 were almost equal in classic and follicular PTC, all of the TCV-positive cases expressed the RET/PTC3 rearrangement. These findings prompted us to compare RET/PTC3 and RET/PTC1 in an in vitro thyroid model system. We have expressed the two oncogenes in PC Cl 3 rat thyroid epithelial cells and found that RET/PTC3 is endowed with a strikingly more potent mitogenic effect than RET/PTC1. Mechanistically, this difference correlated with an increased signaling activity of RET/PTC3. In conclusion, we postulate that the correlation between the RET/PTC rearrangement type and the aggressiveness of human PTC is related to the efficiency with which the oncogene subtype delivers mitogenic signals to thyroid cells.


Nature Clinical Practice Endocrinology & Metabolism | 2006

Drug Insight: small-molecule inhibitors of protein kinases in the treatment of thyroid cancer

Massimo Santoro; Francesca Carlomagno

Molecular targeting of protein kinases is a new paradigm in the treatment of cancer. The clinical efficacy of low-molecular weight inhibitors of ABL, stem-cell growth-factor receptor, and the epidermal growth factor receptor in different tumor types is witness to the power of this approach. The presence of activating mutations of a kinase, or an increased gene copy number, might anticipate tumor responsiveness to its targeting. Thyroid cancer is the most prevalent endocrine malignancy and is frequently associated with the oncogenic conversion of two specific protein kinases, RET and BRAF. Small-molecule inhibitors of both kinases have already reached the clinical testing stage. Protein kinases other than RET and BRAF are also being evaluated for their potential in thyroid-cancer treatment.


British Journal of Cancer | 1993

Involvement of RET oncogene in human tumours: specificity of RET activation to thyroid tumours

Santoro M; N Sabino; Y Ishizaka; T Ushijima; Francesca Carlomagno; A Cerrato; Michele Grieco; C Battaglia; Ml Martelli; C Paulin

Non-thyroid neoplasia were analysed by Southern blot of genomic DNA and DNA prepared by reverse transcription and amplification by polymerase chain reaction (RT/PCR) for the activation of the RET oncogene. It is known that the rearrangement of RET occurs in about 10%-20% of human thyroid papillary carcinomas. None of 528 non-thyroid tumours showed rearrangement of the RET proto-oncogene, whereas three out of 30 thyroid papillary carcinomas were positive for RET activation. Therefore the activation of RET seems to be a somatic cell mutation specific to human thyroid carcinomas.

Collaboration


Dive into the Francesca Carlomagno's collaboration.

Top Co-Authors

Avatar

Massimo Santoro

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Rosa Marina Melillo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Alfredo Fusco

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Giuliana Salvatore

Maharaja Sayajirao University of Baroda

View shared research outputs
Top Co-Authors

Avatar

Giancarlo Vecchio

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Roberta Visconti

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Teresa Guida

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Gabriella De Vita

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Valentina De Falco

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Gelsy Arianna Lupoli

University of Naples Federico II

View shared research outputs
Researchain Logo
Decentralizing Knowledge