Network


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

Hotspot


Dive into the research topics where Simona Romano is active.

Publication


Featured researches published by Simona Romano.


Cell Death & Differentiation | 2010

Role of FK506-binding protein 51 in the control of apoptosis of irradiated melanoma cells

Simona Romano; Anna D'Angelillo; R Pacelli; Stefania Staibano; E De Luna; Rita Bisogni; E-L Eskelinen; Massimo Mascolo; G Calì; C Arra; Maria Fiammetta Romano

FK506-binding protein 51 (FKBP51) is an immunophilin with isomerase activity, which performs important biological functions in the cell. It has recently been involved in the apoptosis resistance of malignant melanoma. The aim of this study was to investigate the possible role of FKBP51 in the control of response to ionizing radiation (Rx) in malignant melanoma. FKBP51-silenced cells showed reduced clonogenic potential after irradiation compared with non-silenced cells. After Rx, we observed apoptosis in FKBP51-silenced cells and autophagy in non-silenced cells. The FKBP51-controlled radioresistance mechanism involves NF-κB. FKBP51 was required for the activation of Rx-induced NF-κB, which in turn inhibited apoptosis by stimulating X-linked inhibitor of apoptosis protein and promoting authophagy-mediated Bax degradation. Using a tumor–xenograft mouse model, the in vivo pretreatment of tumors with FKBP51-siRNA provoked massive apoptosis after irradiation. Immunohistochemical analysis of 10 normal skin samples and 80 malignant cutaneous melanomas showed that FKBP51 is a marker of melanocyte malignancy, correlating with vertical growth phase and lesion thickness. Finally, we provide evidence that FKBP51 targeting radiosensitizes cancer stem/initiating cells. In conclusion, our study identifies a possible molecular target for radiosensitizing therapeutic strategies against malignant melanoma.


Nature Immunology | 2014

USP15 stabilizes MDM2 to mediate cancer-cell survival and inhibit antitumor T cell responses

Qiang Zou; Jin Jin; Hongbo Hu; Haiyan S. Li; Simona Romano; Yichuan Xiao; Mako Nakaya; Xiaofei Zhou; Xuhong Cheng; Peirong Yang; Guillermina Lozano; Chengming Zhu; Stephanie S. Watowich; Stephen E. Ullrich; Shao Cong Sun

Deubiquitinases (DUBs) are a new class of drug targets, although the physiological function of only few DUBs has been characterized. Here we identified the DUB USP15 as a crucial negative regulator of T cell activation. USP15 stabilized the E3 ubiquitin ligase MDM2, which in turn negatively regulated T cell activation by targeting the degradation of the transcription factor NFATc2. USP15 deficiency promoted T cell activation in vitro and enhanced T cell responses to bacterial infection and tumor challenge in vivo. USP15 also stabilized MDM2 in cancer cells and regulated p53 function and cancer-cell survival. Our results suggest that inhibition of USP15 may both induce tumor cell apoptosis and boost antitumor T cell responses.


Cell Death and Disease | 2013

FK506 binding protein 51 positively regulates melanoma stemness and metastatic potential

Simona Romano; Stefania Staibano; Adelaide Greco; Arturo Brunetti; Giovanna Nappo; Gennaro Ilardi; Rosanna Martinelli; Antonio Sorrentino; A Di Pace; Massimo Mascolo; Rita Bisogni; M Scalvenzi; Bruno Alfano; Maria Fiammetta Romano

Melanoma is the most aggressive skin cancer; there is no cure in advanced stages. Identifying molecular participants in melanoma progression may provide useful diagnostic and therapeutic tools. FK506 binding protein 51 (FKBP51), an immunophilin with a relevant role in developmental stages, is highly expressed in melanoma and correlates with aggressiveness and therapy resistance. We hypothesized a role for FKBP51 in melanoma invasive behaviour. FKBP51 promoted activation of epithelial-to-mesenchymal transition (EMT) genes and improved melanoma cell migration and invasion. In addition, FKBP51 induced some melanoma stem cell (MCSC) genes. Purified MCSCs expressed high EMT genes levels, suggesting that genetic programs of EMT and MCSCs overlap. Immunohistochemistry of samples from patients showed intense FKBP51 nuclear signal and cytoplasmic positivity for the stem cell marker nestin in extravasating melanoma cells and metastatic brains. In addition, FKBP51 targeting by small interfering RNA (siRNA) prevented the massive metastatic substitution of liver and lung in a mouse model of experimental metastasis. The present study provides evidence that the genetic programs of cancer stemness and invasiveness overlap in melanoma, and that FKBP51 plays a pivotal role in sustaining such a program.


Cardiovascular Research | 2008

FK506 can activate transforming growth factor-β signalling in vascular smooth muscle cells and promote proliferation

Arturo Giordano; Simona Romano; Maria Mallardo; Anna D'Angelillo; Gaetano Calì; Nicola Corcione; Paolo Ferraro; Maria Fiammetta Romano

AIMS FK506-binding protein (FKBP) 12 is an inhibitor of transforming growth factor (TGF)-beta type I receptors. Several lines of evidence support the view that TGF-beta stimulates vascular smooth muscle cell (VSMC) proliferation and matrix accumulation. We investigated the effect of FK506, also known as tacrolimus, on cellular proliferation and on matrix protein production in human VSMCs. METHODS AND RESULTS We measured cell proliferation with flow cytometry using BrdU incorporation and fluorimetrically by measuring DNA concentration with Hoechst 33258. Western blot assay of whole-cell lysates was used to measure the levels of signalling proteins involved in proliferative pathways, in particular beta-catenin, pErk, pAkt, pmTOR, and cyclin D1. Collagen synthesis was also investigated by Western blotting. The TGF-beta signal was studied by both Western blotting and confocal microscopy. We used the SiRNA technique for FKBP12 gene silencing. Our results show that FK506 stimulates VSMC proliferation and collagen type I production. FK506 enhanced beta-catenin levels and activated the extracellular signal-regulated kinase, Akt, and mammalian target of rapamycin kinase, which are important effectors of proliferation. Accordingly, cyclin D1 expression was increased. We also demonstrate that FK506 activates the TGF-beta signal in VSMCs and that, through this mechanism, it stimulates cell proliferation. CONCLUSION FK506 can act as a growth factor for VSMCs.


Current Opinion in Pharmacology | 2011

FKBP51 and the NF-κB regulatory pathway in cancer

Simona Romano; Massimo Mallardo; Maria Fiammetta Romano

Constitutive activation of NF-κB occurs in a significant percentage of human cancers. Genetic abnormalities of tumors often enhance normal NF-κB signaling. Chronic inflammation is also associated with constitutive NF-κB activation and increases the risk of cancer. Aberrant NF-κB activation favors cellular transformation, sustains cancer survival, and contributes to tumor invasion. Strategies to inhibit NF-κB represent a promising therapeutic option against cancer. In the last decade, several studies point to the large immunophilin FKBP51 as an important element for the control of NF-κB activation in human neoplasia. This article is an overview of the causes of aberrant NF-κB regulation in cancer and highlights recent papers that implicate FKBP51 in such deregulation.


Clinical and translational medicine | 2014

FKBP51 increases the tumour-promoter potential of TGF-beta

Simona Romano; Anna D’Angelillo; Paolo D’Arrigo; Stefania Staibano; Adelaide Greco; Arturo Brunetti; Massimiliano Scalvenzi; Rita Bisogni; Iris Scala; Maria Fiammetta Romano

BackgroundFKBP51 (FKBP5 Official Symbol) is a large molecular weight component of the family of FK506 binding proteins (FKBP). In recent years, research studies from our laboratory highlighted functions for FKBP51 in the control of apoptosis and melanoma progression. FKBP51 expression correlated with the invasiveness and aggressiveness of melanoma. Since a role for TGF-β in the enhanced tumorigenic potential of melanoma cells is widely described, we hypothesized a cooperative effect between FKBP51 and TGF-β in melanoma progression.MethodsSAN and A375 melanoma cell lines were utilized for this study. Balb/c IL2γ NOD SCID served to assess the ability to colonize organs and metastasize of different cell lines, which was evaluated by in vivo imaging. Realtime PCR and western blot served for measurement of mRNA and protein expression, respectively.ResultsBy comparing the metastatic potential of two melanoma cell lines, namely A375 and SAN, we confirmed that an increased capability to colonize murine organs was associated with increased levels of FKBP51. A375 melanoma cell line expressed FKBP51 mRNA levels 30-fold higher in comparison to the SAN mRNA level and appeared more aggressive than SAN melanoma cell line in an experimental metastasis model. In addition, A375 expressed, more abundantly than SAN, the TGF-β and the pro angiogenic TGF-β receptor type III (TβRIII) factors. FKBP51 silencing produced a reduction of TGF-β and TβRIII gene expression in A375 cell line, in accordance with previous studies. We found that the inducing effect of TGF-β on Sparc and Vimentin expression was impaired in condition of FKBP51 depletion, suggesting that FKBP51 is an important cofactor in the TGF-β signal. Such a hypothesis was supported by co immunoprecipitation assays, showing that FKBP51 interacted with either Smad2,3 and p300. In normal melanocytes, FKBP51 potentiated the effect of TGF-β on N-cadherin expression and conferred a mesenchymal-like morphology to such round-shaped cells.ConclusionsOverall, our findings show that FKBP51 enhances some pro oncogenic functions of TGF-β, suggesting that FKBP51-overexpression may help melanoma to take advantage of the tumor promoting activities of the cytokine.


Cell Death and Disease | 2010

FK506-binding protein 51 is a possible novel tumoral marker

Simona Romano; A D'Angelillo; Stefania Staibano; Gennaro Ilardi; Maria Fiammetta Romano

Dear Editor, FK506-binding protein (FKBP) 51 is a cochaperone, which belongs to the immunophilin family, a group of proteins with peptidyl-prolyl isomerase activity. FKBPs regulate several biological processes through protein–protein interaction.1 In particular, FKBP51 is a component of the steroid receptor complex, with a role in steroid resistance;2 moreover, it is involved in NF-κB activation because of its isomerase activity, which is essential for the function of subunit-α in the IκB kinase complex.3 According to Baughman et al.,4 FKBP51 is abundantly expressed in lymphocytes and in several other tissues, but it is expressed at low levels in the pancreas, spleen, and stomach. There is increasing evidence of an association of FKBP51 hyperexpression with cancer6, 7, 8 and a relevant role of this protein in sustaining cell growth,5 malignancy, and resistance to therapy.6, 7, 8 An immunohistochemistry study of expression of FKBP51 in 50 tumoral samples acquired from our pathology section, including breast, lung, pancreas, ovary, and prostate (10 samples for each tumor), and a comparable number of normal tissue samples showed an intense signal in 38 out of 50 tumors analyzed, whereas normal tissues of the same histotypes showed a weak/absent immunohistochemical signal. The 12 tumor samples with low/negative immunohistochemistry were the 10 breast cancer samples and 2 out of 10 pancreatic tumors. Interestingly, these two pancreatic tumors belonged to the well-differentiated histotype (G1). Figure 1a (upper panel) shows representative immunohistochemical findings. Measurement of FKBP51 mRNA levels in deparaffinized tissues using real-time PCR confirmed the immunohistochemistry results (representative results in Figure 1a, lower panel). Taken together, these findings support the hypothesis that FKBP51 is a promising novel tumoral marker. The association of FKBP51 overexpression with cancer is in line with the rapidly emerging concept that NF-κB drives tumorigenesis in the most common genetic alterations associated with cancer.9, 10 Figure 1 (a) (Upper panel) FKBP51 immunochemical staining of normal and neoplastic tissues. Serial sections of 4 μm from routinely formalin-fixed, paraffin-embedded blocks were cut and mounted on poly--lysine-coated glass slides. (Lower panel) ... Recently, it has been found, in tumor cell lines, that FKBP51 acted as a scaffold to facilitate the interaction between Akt and PH domain leucine-rich repeat protein phosphatase, which mediates dephosphorylation of pAkt (S473).11 This raised the question whether FKBP51 may work out as a tumor suppressor by deactivating Akt. As we previously found that intratumoral injection of FKBP51 siRNA followed by irradiation produced extensive apoptosis in melanoma xenografts,8 we investigated the effect of FKBP51 downmodulation on pAkt (S473) levels in our mouse model of melanoma, in both xenografts and locoregional lymph nodes. Histological examination showed that the mouse lymphatic tissue was not metastatic. Figure 1b shows a representative outcome. On the basis of these results, pAkt (S473) levels were not enhanced with downmodulation of FKBP51 in normal or cancerous tissue. This observation suggests that pathways upstream from Akt, which are often deregulated in cancer, control activated-Akt levels. This hypothesis is supported by the findings that leukemic lymphocytes often display higher levels of pAkt (S473) in comparison with normal lymphocytes (Figure 1c) even in the presence of similar or higher FKBP51 levels. To assess whether an inverse correlation subsisted between FKBP51 and pAkt (S473) levels, we used western blot to measure these levels in samples of primary lymphatic leukemia and normal peripheral blood lymphocytes. Expression levels were quantified by densitometry. Spearmans ρ correlation did not indicate any relationship between the two variables (Figure 1d; P=0.788, left; P=0.199, right). Taken together, these findings do not support an essential role of FKBP51 as a factor that controls the phosphorylation status of Akt inside the cell, thereby weakening the hypothesis that the protein may work out as tumor suppressor. In conclusion, although FKBP51 function is not yet fully elucidated, however, there are clear data suggesting that this immunophilin is often hyperexpressed in tumors and has an active role in pre-neoplastic disorders5 and cancer.6, 7, 8


Biochimie | 2013

Markers of mitochondrial dysfunction during the diclofenac-induced apoptosis in melanoma cell lines

Francesco Albano; Alessandro Arcucci; Giuseppina Granato; Simona Romano; Stefania Montagnani; Emmanuele De Vendittis; Maria Rosaria Ruocco

Melanoma is an aggressive cutaneous cancer, whose incidence is growing in recent years, especially in the younger population. The favorable therapy for this neoplasm consists in its early surgical excision; otherwise, in case of late diagnosis, melanoma becomes very refractory to any conventional therapy. Nevertheless, the acute inflammatory response occurring after excision of the primary melanoma can affect the activation and/or regulation of melanoma invasion and metastasis. Nonsteroidal anti-inflammatory drugs (NSAIDs), widely employed in clinical therapy as cyclooxygenase inhibitors, also display a cytotoxic effect on some cancer cell lines; therefore, their possible usage in combination with conventional chemo- and radio-therapies of tumors is being considered. In particular, diclofenac, one of the most common NSAIDs, displays its anti-proliferative effect in many tumor lines, through an alteration of the cellular redox state. In this study, the possible anti-neoplastic potential of diclofenac on the human melanoma cell lines A2058 and SAN was investigated, and a comparison was made with the results obtained from the nonmalignant fibroblast cell line BJ-5ta. Either in A2058 or SAN, the diclofenac treatment caused typical apoptotic morphological changes, as well as an increase of the number of sub-diploid nuclei; conversely, the same treatment on BJ-5ta had only a marginal effect. The observed decrease of Bcl-2/Bax ratio and a parallel increase of caspase-3 activity confirmed the pro-apoptotic role exerted by diclofenac in melanoma cells; furthermore, the drug provoked an increase of the ROS levels, a decrease of mitochondrial superoxide dismutase (SOD2), the cytosolic translocation of both SOD2 and cytochrome c, and an increase of caspase-9 activity. Finally, the cytotoxic effect of diclofenac was amplified, in melanoma cells, by the silencing of SOD2. These data improve the knowledge on the effects of diclofenac and suggest that new anti-neoplastic treatments should be based on the central role of mitochondrion in cancer development; under this concern, the possible involvement of SOD2 as a novel target could be considered.


Nucleic Acids Research | 2015

FKBP51 employs both scaffold and isomerase functions to promote NF-κB activation in melanoma

Simona Romano; Yichuan Xiao; Mako Nakaya; Anna D'Angelillo; Mikyoung Chang; Jin Jin; Felix Hausch; Mariorosario Masullo; Xixi Feng; Maria Fiammetta Romano; Shao Cong Sun

Melanoma is the most aggressive skin cancer; its prognosis, particularly in advanced stages, is disappointing largely due to the resistance to conventional anticancer treatments and high metastatic potential. NF-κB constitutive activation is a major factor for the apoptosis resistance of melanoma. Several studies suggest a role for the immunophilin FKBP51 in NF-κB activation, but the underlying mechanism is still unknown. In the present study, we demonstrate that FKBP51 physically interacts with IKK subunits, and facilitates IKK complex assembly. FKBP51-knockdown inhibits the binding of IKKγ to the IKK catalytic subunits, IKK-α and -β, and attenuates the IKK catalytic activity. Using FK506, an inhibitor of the FKBP51 isomerase activity, we found that the IKK-regulatory role of FKBP51 involves both its scaffold function and its isomerase activity. Moreover, FKBP51 also interacts with TRAF2, an upstream mediator of IKK activation. Interestingly, both FKBP51 TPR and PPIase domains are required for its interaction with TRAF2 and IKKγ, whereas only the TPR domain is involved in interactions with IKKα and β. Collectively, these results suggest that FKBP51 promotes NF-κB activation by serving as an IKK scaffold as well as an isomerase. Our findings have profound implications for designing novel melanoma therapies based on modulation of FKBP51.


Current Medicinal Chemistry | 2011

The Emerging Role of Large Immunophilin FK506 Binding Protein 51 in Cancer

Simona Romano; Antonio Sorrentino; A. L. Di Pace; Giovanna Nappo; C. Mercogliano; Maria Fiammetta Romano

FK506 binding protein 51 (FKBP51) is an immunophilin physiologically expressed in lymphocytes. Very recently, aberrant expression of this protein was found in melanoma; FKBP51 expression correlates with melanoma aggressiveness and is maximal in metastatic lesions. FKBP51 promotes NF-κB activation and is involved in the resistance to genotoxic agents, including anthracyclines and ionizing radiation. FKBP51 is a cochaperone with peptidyl-prolyl isomerase activity that regulates several biological processes through protein-protein interaction. There is increasing evidence that FKBP51 hyperexpression is associated with cancer and this protein has a relevant role in sustaining cell growth, malignancy, and resistance to therapy. There is also evidence that FKBP ligands are potent anticancer agents, in addition to their immunosuppressant activity. In particular, rapamycin and its analogs have shown antitumor activity across a variety of human cancers in clinical trials. Although, classically, rapamycin actions are ascribed to inhibition of mTOR, recent studies indicate FKBP51 is also an important molecular determinant of the drug’s anticancer activity. The aim of this article is to review the functions of FKBP51, especially in view of the recent findings that this protein is a potential oncogene when deregulated and a candidate target for signaling therapies against cancer.

Collaboration


Dive into the Simona Romano's collaboration.

Top Co-Authors

Avatar

Maria Fiammetta Romano

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Rita Bisogni

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Stefania Staibano

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Anna D'Angelillo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Gennaro Ilardi

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Massimo Mascolo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Paolo D'Arrigo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Arturo Giordano

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Michele Russo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Antonio Sorrentino

Istituto Superiore di Sanità

View shared research outputs
Researchain Logo
Decentralizing Knowledge