Network


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

Hotspot


Dive into the research topics where Francesco Romeo is active.

Publication


Featured researches published by Francesco Romeo.


Cell Cycle | 2013

GLS2 is transcriptionally regulated by p73 and contributes to neuronal differentiation

Tania Velletri; Francesco Romeo; Paola Tucci; Angelo Peschiaroli; Margherita Annicchiarico-Petruzzelli; Maria Victoria Niklison-Chirou; Ivano Amelio; Richard A. Knight; Tak W. Mak; Gerry Melino; Massimiliano Agostini

The amino acid Glutamine is converted into Glutamate by a deamidation reaction catalyzed by the enzyme Glutaminase (GLS). Two isoforms of this enzyme have been described, and the GLS2 isoform is regulated by the tumor suppressor gene p53. Here, we show that the p53 family member TAp73 also drives the expression of GLS2. Specifically, we demonstrate that TAp73 regulates GLS2 during retinoic acid-induced terminal neuronal differentiation of neuroblastoma cells, and overexpression or inhibition of GLS2 modulates neuronal differentiation and intracellular levels of ATP. Moreover, inhibition of GLS activity, by removing Glutamine from the growth medium, impairs in vitro differentiation of cortical neurons. Finally, expression of GLS2 increases during mouse cerebellar development. Although, p73 is dispensable for the in vivo expression of GLS2, TAp73 loss affects GABA and Glutamate levels in cortical neurons. Together, these findings suggest a role for GLS2 acting, at least in part, downstream of p73 in neuronal differentiation and highlight a possible role of p73 in regulating neurotransmitter synthesis.


Breast Cancer Research and Treatment | 2014

Tamoxifen through GPER upregulates aromatase expression: a novel mechanism sustaining tamoxifen-resistant breast cancer cell growth

Stefania Catalano; Cinzia Giordano; Salvatore Panza; Francesca Chemi; Daniela Bonofiglio; Marilena Lanzino; Pietro Rizza; Francesco Romeo; Suzanne A. W. Fuqua; Marcello Maggiolini; Sebastiano Andò; Ines Barone

Tamoxifen resistance is a major clinical challenge in breast cancer treatment. Aromatase inhibitors are effective in women who progressed or recurred on tamoxifen, suggesting a role of local estrogen production by aromatase in driving tamoxifen-resistant phenotype. However, the link between aromatase activity and tamoxifen resistance has not yet been reported. We investigated whether long-term tamoxifen exposure may affect aromatase activity and/or expression, which may then sustain tamoxifen-resistant breast cancer cell growth. We employed MCF-7 breast cancer cells, tamoxifen-resistant MCF-7 cells (MCF-7 TR1 and TR2), SKBR-3 breast cancer cells, cancer-associated fibroblasts (CAFs1 and CAFs2). We used tritiated-water release assay, realtime-RT-PCR, and immunoblotting analysis for evaluating aromatase activity and expression; anchorage-independent assays for growth; reporter-gene, electrophoretic-mobility-shift, and chromatin-immunoprecipitation assays for promoter activity studies. We demonstrated an increased aromatase activity and expression, which supports proliferation in tamoxifen-resistant breast cancer cells. This is mediated by the G-protein-coupled receptor GPR30/GPER, since knocking-down GPER expression or treatment with a GPER antagonist reversed the enhanced aromatase levels induced by long-term tamoxifen exposure. The molecular mechanism was investigated in ER-negative, GPER/aromatase-positive SKBR3 cells, in which tamoxifen acts as a GPER agonist. Tamoxifen treatment increased aromatase promoter activity through an enhanced recruitment of c-fos/c-jun complex to AP-1 responsive elements located within the promoter region. As tamoxifen via GPER induced aromatase expression also in CAFs, this pathway may be involved in promoting aggressive behavior of breast tumors in response to tamoxifen treatment. Blocking estrogen production and/or GPER signaling activation may represent a valid option to overcome tamoxifen-resistance in breast cancers.


Cell Cycle | 2013

The estrogen receptor α is the key regulator of the bifunctional role of FoxO3a transcription factor in breast cancer motility and invasiveness

Diego Sisci; Pamela Maris; Maria Grazia Cesario; Wanda Anselmo; Roberta Coroniti; Giovanna Elvi Trombino; Francesco Romeo; Aurora Ferraro; Marilena Lanzino; Saveria Aquila; Marcello Maggiolini; Loredana Mauro; Catia Morelli; Sebastiano Andò

The role of the Forkhead box class O (FoxO)3a transcription factor in breast cancer migration and invasion is controversial. Here we show that FoxO3a overexpression decreases motility, invasiveness, and anchorage-independent growth in estrogen receptor α-positive (ERα+) cancer cells while eliciting opposite effects in ERα-silenced cells and in ERα-negative (ERα−) cell lines, demonstrating that the nuclear receptor represents a crucial switch in FoxO3a control of breast cancer cell aggressiveness. In ERα+ cells, FoxO3a-mediated events were paralleled by a significant induction of Caveolin-1 (Cav1), an essential constituent of caveolae negatively associated to tumor invasion and metastasis. Cav1 induction occurs at the transcriptional level through FoxO3a binding to a Forkhead responsive core sequence located at position −305/−299 of the Cav1 promoter. 17β-estradiol (E2) strongly emphasized FoxO3a effects on cell migration and invasion, while ERα and Cav1 silencing were able to reverse them, demonstrating that both proteins are pivotal mediators of these FoxO3a controlled processes. In vivo, an immunohistochemical analysis on tissue sections from patients with ERα+ or ERα− invasive breast cancers or in situ ductal carcinoma showed that nuclear FoxO3a inversely (ERα+) or directly (ERα−) correlated with the invasive phenotype of breast tumors. In conclusion, FoxO3a role in breast cancer motility and invasion depends on ERα status, disclosing a novel aspect of the well-established FoxO3a/ERα interplay. Therefore FoxO3a might become a pursuable target to be suitably exploited in combination therapies either in ERα+ or ERα− breast tumors.


Clinical Cancer Research | 2008

BRCA1 5083del19 mutant allele selectively up-regulates periostin expression in vitro and in vivo

Barbara Quaresima; Francesco Romeo; Maria Concetta Faniello; Maddalena Di Sanzo; Chang Gong Liu; Annamaria Lavecchia; Cristian Taccioli; Eugenio Gaudio; Francesco Baudi; Francesco Trapasso; Carlo M. Croce; Giovanni Cuda; Francesco Costanzo

Purpose: The aim of this study was to explore the gene expression pattern produced by the cancer-associated BRCA1 5083del19 founder mutation by using a microarray analysis. Such a mutation, identified in a subset of familial breast cancer patients, involves a deletion at the 3′ end of the BRCA1 messenger leading, in the mature protein, to the ablation of the BRCT tandem domain. Experimental Design: We generated HeLa cells stably expressing both exogenous wild-type (HeLa/wtBRCA1), used as a control, and 5083del19 BRCA1 (HeLa/5083del19BRCA1) alleles; gene chips were then used to investigate any changes in the transcription profile induced by the 5083del19 BRCA1 mutant compared with controls. Results: Among the genes showing perturbation of their expression, periostin was found to be up-regulated in HeLa/5083del19BRCA1 cells to an extent of 72-fold versus HeLa/pcDNA3.1/empty and 76-fold versus HeLa/wtBRCA1 cells. This finding was validated both in vitro in breast cancer cell lines harboring mutations of BRCA1 and in vivo by immunohistochemistry of breast cancer specimens bearing the 5083del19 BRCA1 mutation as well as by Western blot analysis of sera obtained from patients and healthy carriers of the same mutation. Conclusions: Our results suggest that periostin overexpression, whose product is released from cells in the extracellular fluids, might be a potential marker for early cancer detection in a specific subset of hereditary breast carcinomas triggered by cancer-associated BRCA1 mutations that affect the BRCT tandem domain.


Gene | 2014

Identification of H ferritin-dependent and independent genes in K562 differentiating cells by targeted gene silencing and expression profiling

Roberta Misaggi; Maddalena Di Sanzo; Carlo Cosentino; Heather M. Bond; Domenica Scumaci; Francesco Romeo; Claudia Stellato; Giorgio Giurato; Alessandro Weisz; Barbara Quaresima; Tullio Barni; Francesco Amato; Giuseppe Viglietto; Giovanni Morrone; Giovanni Cuda; Maria Concetta Faniello; Francesco Costanzo

Ferritin is best known as the key molecule in intracellular iron storage, and is involved in several metabolic processes such as cell proliferation, differentiation and neoplastic transformation. We have recently demonstrated that the shRNA silencing of the ferritin heavy subunit (FHC) in a melanoma cell line is accompanied by a consistent modification of gene expression pattern leading to a reduced potential in terms of proliferation, invasiveness, and adhesion ability of the silenced cells. In this study we sought to define the repertoire of genes whose expression might be affected by FHC during the hemin-induced differentiation of the erythromyeloid cell line K562. To this aim, gene expression profiling was performed in four different sets of cells: i) wild type K562; ii) sh-RNA FHC-silenced K562; iii) hemin-treated wild-type K562; and iv) hemin-treated FHC-silenced K562. Statistical analysis of the gene expression data, performed by two-factor ANOVA, identified three distinct classes of transcripts: a) Class 1, including 657 mRNAs whose expression is modified exclusively during hemin-induced differentiation of K562 cells, independently from the FHC relative amounts; b) Class 2, containing a set of 70 mRNAs which are consistently modified by hemin and FHC-silencing; and c) Class 3, including 128 transcripts modified by FHC-silencing but not by hemin. Our data indicate that FHC may function as a modulator of gene expression during erythroid differentiation and add new findings to the knowledge of the complex gene network modulated during erythroid differentiation.


Clinical Cancer Research | 2016

Expression and Function of Phosphodiesterase Type 5 in Human Breast Cancer Cell Lines and Tissues: Implications for Targeted Therapy

Stefania Catalano; Antonella Campana; Cinzia Giordano; Balázs Győrffy; Roberta Tarallo; Antonio Rinaldi; Giuseppina Bruno; Aurora Ferraro; Francesco Romeo; Marilena Lanzino; Fabio Naro; Daniela Bonofiglio; Sebastiano Andò; Ines Barone

Purpose: By catalyzing cGMP hydrolysis, phosphodiesterase (PDE) 5 is a critical regulator of its concentration and effects in different (patho)physiologic processes, including cancers. As PDE5 is a known druggable target, we investigated the clinical significance of its expression in breast cancer and the underlying mechanisms by which it may contribute to tumor progression. Experimental Design: PDE5 expression was evaluated in seven breast cancer cell lines by RT-PCR and immunoblotting. To examine the impact of PDE5 on cancer phenotype, MCF-7 cells expressing lower levels of the enzyme were engineered to stably overexpress PDE5. Proliferation was evaluated by MTT assays, motility and invasion by wound-healing/transmigration/invasion assays, transcriptome-profiling by RNA-sequencing, and Rho GTPase signaling activation by GST-pulldown assays and immunoblotting. Clinical relevance was investigated by IHC on tissues and retrospective studies from METABRIC cohort. Results: PDE5 is differentially expressed in each molecular subtype of both breast cancer cell lines and tissues, with higher levels representing a startling feature of HER2-positive and triple-negative breast cancers. A positive correlation was established between elevated PDE5 levels and cancers of high histologic grade. Higher PDE5 expression correlated with shorter patient survival in retrospective analyses. On molecular level, stable PDE5 overexpression in Luminal-A–like MCF-7 cells resulted in enhanced motility and invasion through Rho GTPase signaling activation. Treatment of PDE5-stable clones with selective ROCK or PDE5 inhibitors completely restored the less motile and weak invasive behavior of control vector cells. Conclusions: PDE5 expression enhances breast cancer cell invasive potential, highlighting this enzyme as a novel prognostic candidate and an attractive target for future therapy in breast cancers. Clin Cancer Res; 22(9); 2271–82. ©2015 AACR.


The International Journal of Biochemistry & Cell Biology | 2011

BRCA1 is required for hMLH1 stabilization following doxorubicin-induced DNA damage

Francesco Romeo; Lucia Falbo; Maddalena Di Sanzo; Roberta Misaggi; Maria Concetta Faniello; Giuseppe Viglietto; Giovanni Cuda; Francesco Costanzo; Barbara Quaresima

Human DNA mismatch repair (MMR) is involved in the removal of DNA base mismatches that arise either during DNA replication or are caused by DNA damage. In this study, we show that the activation of the MMR component hMLH1 in response to doxorubicin (DOX) treatment requires the presence of BRCA1 and that this phenomenon is mediated by an ATM/ATR dependent phosphorylation of the hMLH1 Ser-406 residue. BRCA1 is an oncosuppressor protein with a central role in the DNA damage response and it is a critical component of the ATM/ATR mediated checkpoint signaling. Starting from a previous finding in which we demonstrated that hMLH1 is able to bind to BRCA1, in this study we asked whether BRCA1 might be the bridge for ATM/ATR dependent phosphorylation of the hMLH1 molecular partner. We found that: (i) the negative modulation of BRCA1 expression is able to produce a remarkable reversal of hMLH1 stabilization, (ii) BRCA1 is required for post-translational modification produced by DOX treatment on hMLH1 which is, in turn, attributed to the ATM/ATR activity, (iii) the serine 406 phosphorylatable residue is critical for hMLH1 activation by ATM/ATR via BRCA1. Taken together, our data lend support to the hypothesis suggesting an important role of this oncosuppressor as a scaffold or bridging protein in DNA-damage response signaling via downstream phosphorylation of the ATM/ATR substrate hMLH1.


Journal of Proteome Research | 2011

H ferritin gene silencing in a human metastatic melanoma cell line: a proteomic analysis.

Maddalena Di Sanzo; Marco Gaspari; Roberta Misaggi; Francesco Romeo; Lucia Falbo; Carmela De Marco; Valter Agosti; Barbara Quaresima; Tullio Barni; Giuseppe Viglietto; Martin R. Larsen; Giovanni Cuda; Francesco Costanzo; Maria Concetta Faniello

Ferritin, the major intracellular iron-storage protein, is made of 24 subunits of two types, H and L. Besides regulating intracellular iron homeostasis, it has been found that ferritin, in particular the H subunit (FHC), is involved in different biological events such as cell differentiation and pathologic states (i.e., neurodegeneration and cancer). This study is aimed at investigating the whole-cell proteome of FHC-expressing and sh-RNA-silenced human metastatic melanoma cells (MM07(m)) in the attempt to identify and classify the highest number of proteins directly or indirectly controlled by the FHC. We identified about 200 differentially expressed proteins and classified them in clusters on the basis of their functions, as proteins involved in metabolic processes, cell adhesion, migration, and proliferation processes. Some of them have captured our attention because of their involvement in metabolic pathways related to tumor progression and metastasis. In vitro assays confirmed that the FHC-silenced MM07(m) cells are characterized by a decreased growth activity, a reduced invasiveness, and a reduced cell adhesion capability. Moreover, nude mice (CD1 nu/nu), subcutaneously injected with FHC-silenced MM07(m) cells, showed a remarkable 4-fold reduction of their tumor growth capacity compared to those who received the FHC-unsilenced MM07(m) counterpart. In conclusion, these data indicate that gene silencing technology, coupled to proteomic analysis, is a powerful tool for a better understanding of H ferritin signaling pathways and lend support to the hypothesis that specific targeting of this gene might be an attractive and potentially effective strategy for the management of metastatic melanoma.


Gene | 2011

Negative transcriptional regulation of the human periostin gene by YingYang-1 transcription factor.

Francesco Romeo; Lucia Falbo; M. Di Sanzo; Roberta Misaggi; Maria Concetta Faniello; Tullio Barni; Giovanni Cuda; Giuseppe Viglietto; C. Santoro; Barbara Quaresima; Francesco Costanzo

Periostin (POSTN), an osteoblast-specific secreted protein known to be associated with cell adhesion activity for bone formation and development by the epithelial cell-derived tumors, leads to a significant enhancement in angiogenesis and tumorigenesis. At present, little is known about the mechanisms underlying its transcriptional control either in physiological or neoplastic conditions. In this study we demonstrate that the ability of the human POSTN promoter to drive transcription mostly depends on the activity of YingYang-1 (YY1) zinc finger transcription factor. YY1, whose regulatory role in biology includes, besides transcriptional control, also chromatin remodeling, DNA damage repair and tumorigenesis, acts as a strong negative modulator of the POSTN expression. We retain that the identification of the functional role of YY1 in the transcriptional control of the human POSTN gene adds new insights in the studies focused on gene expression in normal and transformed cells.


Cell Cycle | 2015

Erratum to GLS2 is transcriptionally regulated by p73 and contributes to neuronal differentiation (Cell Cycle, 12, 22, 3564-3573, 10.4161/cc.26771)

Tania Velletri; Francesco Romeo; Paola Tucci; Angelo Peschiaroli; Margherita Annicchiarico-Petruzzelli; Maria Victoria Niklison-Chirou; Ivano Amelio; Richard A. Knight; Tak W. Mak; Gerry Melino; Massimiliano Agostini

Article title: GLS2 is transcriptionally regulated by p73 and contributes to neuronal differentiation Authors: Tania Velletri, Francesco Romeo, Paola Tucci, Angelo Peschiaroli, Margherita Annicchiarico-Petruzzelli, Maria Niklison-Chirou, Ivano Amelio, Richard Knight, Tak Mak, Gerry Melino, and Massimiliano Agostini Journal: Cell Cycle Bibliometrics: Volume 12, Issue 22, Pages 3564–3573 DOI: 10.4161/cc.26771

Collaboration


Dive into the Francesco Romeo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ines Barone

University of Calabria

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerry Melino

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge