Network


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

Hotspot


Dive into the research topics where Maria Grazia Masiello is active.

Publication


Featured researches published by Maria Grazia Masiello.


Journal of Pineal Research | 2014

Melatonin down-regulates MDM2 gene expression and enhances p53 acetylation in MCF-7 cells.

Sara Proietti; Alessandra Cucina; Gabriella Dobrowolny; Fabrizio D'Anselmi; Simona Dinicola; Maria Grazia Masiello; Alessia Pasqualato; Alessandro Palombo; Veronica Morini; Russel J. Reiter; Mariano Bizzarri

Compelling evidence demonstrated that melatonin increases p53 activity in cancer cells. p53 undergoes acetylation to be stabilized and activated for driving cells destined for apoptosis/growth inhibition. Over‐expression of p300 induces p53 acetylation, leading to cell growth arrest by increasing p21 expression. In turn, p53 activation is mainly regulated in the nucleus by MDM2. MDM2 also acts as E3 ubiquitin ligase, promoting the proteasome‐dependent p53 degradation. MDM2 entry into the nucleus is finely tuned by two different modulations: the ribosomal protein L11, acts by sequestering MDM2 in the cytosol, whereas the PI3K‐AkT‐dependent MDM2 phosphorylation is mandatory for MDM2 translocation across the nuclear membrane. In addition, MDM2‐dependent targeting of p53 is regulated in a nonlinear fashion by MDM2/MDMX interplay. Melatonin induces both cell growth inhibition and apoptosis in MCF7 breast cancer cells. We previously reported that this effect is associated with reduced MDM2 levels and increased p53 activity. Herein, we demonstrated that melatonin drastically down‐regulates MDM2 gene expression and inhibits MDM2 shuttling into the nucleus, given that melatonin increases L11 and inhibits Akt‐PI3K‐dependent MDM2 phosphorylation. Melatonin induces a 3‐fold increase in both MDMX and p300 levels, decreasing simultaneously Sirt1, a specific inhibitor of p300 activity. Consequently, melatonin‐treated cells display significantly higher values of both p53 and acetylated p53. Thus, a 15‐fold increase in p21 levels was observed in melatonin‐treated cancer cells. Our results provide evidence that melatonin enhances p53 acetylation by modulating the MDM2/MDMX/p300 pathway, disclosing new insights for understanding its anticancer effect.


BioMed Research International | 2014

Phenotypic Switch Induced by Simulated Microgravity on MDA-MB-231 Breast Cancer Cells

Maria Grazia Masiello; Alessandra Cucina; Sara Proietti; Palombo A; Pierpaolo Coluccia; D'Anselmi F; Dinicola S; Pasqualato A; Morini; Mariano Bizzarri

Microgravity exerts dramatic effects on cell morphology and functions, by disrupting cytoskeleton and adhesion structures, as well as by interfering with biochemical pathways and gene expression. Impairment of cells behavior has both practical and theoretical significance, given that investigations of mechanisms involved in microgravity-mediated effects may shed light on how biophysical constraints cooperate in shaping complex living systems. By exposing breast cancer MDA-MB-231 cells to simulated microgravity (~0.001 g), we observed the emergence of two morphological phenotypes, characterized by distinct membrane fractal values, surface area, and roundness. Moreover, the two phenotypes display different aggregation profiles and adherent behavior on the substrate. These morphological differences are mirrored by the concomitant dramatic functional changes in cell processes (proliferation and apoptosis) and signaling pathways (ERK, AKT, and Survivin). Furthermore, cytoskeleton undergoes a dramatic reorganization, eventually leading to a very different configuration between the two populations. These findings could be considered adaptive and reversible features, given that, by culturing microgravity-exposed cells into a normal gravity field, cells are enabled to recover their original phenotype. Overall these data outline the fundamental role gravity plays in shaping form and function in living systems.


Toxicology in Vitro | 2013

Nicotine increases survival in human colon cancer cells treated with chemotherapeutic drugs.

Simona Dinicola; Veronica Morini; Pierpaolo Coluccia; Sara Proietti; Fabrizio D’Anselmi; Alessia Pasqualato; Maria Grazia Masiello; Alessandro Palombo; Giorgio De Toma; Mariano Bizzarri; Alessandra Cucina

Cigarette smoking is implicated in the development of colon cancer. Furthermore, nicotine increases cell proliferation and inhibits apoptosis through α7-nicotinic acetylcholine receptor (α7-nAChR) activation in human colon carcinoma cells. An open issue is whether nicotine interfere with colorectal cancer pharmacological treatment, by inhibiting drug-mediated apoptosis. To assess this hypothesis, we evaluated nicotine effect on Caco-2 and HCT-8 colon cancer cells, treated with 5-Fluorouracil (5-FU) and Camptothecin (CPT), chemotherapeutics commonly utilized as adjuvant treatment of colon cancer. Nicotine decreased anti-proliferative and pro-apoptotic effects exerted by chemotherapeutics on both cell lines. These effects partially reverted by exposure to α-bungarotoxin (α-BTX), an inhibitor of α7-nAChR. Nicotine addition to Caco-2 and HCT-8, treated with 5-FU or CPT, decreased the cleavage of substrate of caspase 3 and 7, poly-ADP-ribose polymerase (PARP). Moreover, P-ERK/ERK ratio was modified by nicotine addition to 5-FU and CPT treated cells in an opposite manner. However, when co-administrating PD98059, an ERK phosphorylation inhibitor, an increased apoptosis was observed. In Caco-2 and HCT-8 nicotine reverted 5-FU and CPT apoptotic effects through AKT phosphorylation, as demonstrated by apoptotic increase in presence of LY294002, an AKT phosphorylation inhibitor. Nicotine interfered with colorectal cancer pharmacological treatment in vitro by inhibiting apoptosis induced by chemotherapeutic drugs. Nicotine anti-apoptotic effects were exerted through ERK and AKT pathway activation.


Rend. Fis. Acc. Lincei | 2014

Gravity sensing by cells: mechanisms and theoretical grounds

Mariano Bizzarri; Alessandra Cucina; Alessandro Palombo; Maria Grazia Masiello

Spaceflight technologies have disclosed amazing opportunities to outreach human knowledge and control over the natural world. However, the actual experience of microgravity has become a relevant threat that significantly limits the extent of man permanence in space. Since then, gravity effects on living organisms became a critical field of investigation. Gravity has been proven to affect a wide array of biological functions, interacting at different levels of complexity, from molecules to cells, tissue and the organisms as a whole. However, it is still a matter of investigation if gravity induces direct or indirect effects on cells. The non-equilibrium theory has been proven to explain how biological dissipative structures, like the cytoskeleton, may be sensitive enough to sense gravity change, then transferring the mechano-signal into biochemical pathways. Within that framework, gravity represents an ‘inescapable’ constraint that obliges living beings to adopt only a few configurations among many others. By removing the gravitational field, living structures will be free to recover more degrees of freedom, thus acquiring new phenotypes and new properties. Discoveries on that field are thought to advance our knowledge, providing amazing insights into the biological mechanism underlying physiology as well as many relevant diseases.


BioMed Research International | 2014

Lung Cancer Stem Cell Lose Their Stemness Default State after Exposure to Microgravity

Maria Elena Pisanu; Alessia Noto; Claudia De Vitis; Maria Grazia Masiello; Pierpaolo Coluccia; Sara Proietti; Maria Rosaria Giovagnoli; Alberto Ricci; Enrico Giarnieri; Alessandra Cucina; Gennaro Ciliberto; Mariano Bizzarri; Rita Mancini

Microgravity influences cell differentiation by modifying the morphogenetic field in which stem cells are embedded. Preliminary data showed indeed that stem cells are committed to selective differentiation when exposed to real or simulated microgravity. Our study provides evidence that a similar event occurs when cancer stem cells (CSCs) are cultured in microgravity. In the same time, a significant increase in apoptosis was recorded: those data point out that microgravity rescues CSCs from their relative quiescent state, inducing CSCs to lose their stemness features, as documented by the decrease in ALDH and the downregulation of both Nanog and Oct-4 genes. Those traits were stably acquired and preserved by CSCs when cells were placed again on a 1 g field. Studies conducted in microgravity on CSCs may improve our understanding of the fundamental role exerted by biophysical forces in cancer cell growth and function.


PLOS ONE | 2013

Microenvironment Promotes Tumor Cell Reprogramming in Human Breast Cancer Cell Lines

Fabrizio D’Anselmi; Maria Grazia Masiello; Alessandra Cucina; Sara Proietti; Simona Dinicola; Alessia Pasqualato; Giulia Ricci; Gabriella Dobrowolny; Angela Catizone; Alessandro Palombo; Mariano Bizzarri

The microenvironment drives mammary gland development and function, and may influence significantly both malignant behavior and cell growth of mammary cancer cells. By restoring context, and forcing cells to properly interpret native signals from the microenvironment, the cancer cell aberrant behavior can be quelled, and organization re-established. In order to restore functional and morphological differentiation, human mammary MCF-7 and MDA-MB-231 cancer cells were allowed to grow in a culture medium filled with a 10% of the albumen (EW, Egg White) from unfertilized chicken egg. That unique microenvironment behaves akin a 3D culture and induces MCF-7 cells to produce acini and branching duct-like structures, distinctive of mammary gland differentiation. EW-treated MDA-MB-231 cells developed buds of acini and duct-like structures. Both MCF-7 and MDA-MB-231 cells produced β-casein, a key milk component. Furthermore, E-cadherin expression was reactivated in MDA-MB-231 cells, as a consequence of the increased cdh1 expression; meanwhile β-catenin – a key cytoskeleton component – was displaced behind the inner cell membrane. Such modification hinders the epithelial-mesenchymal transition in MDA-MB-231 cells. This differentiating pathway is supported by the contemporary down-regulation of canonical pluripotency markers (Klf4, Nanog). Given that egg-conditioned medium behaves as a 3D-medium, it is likely that cancer phenotype reversion could be ascribed to the changed interactions between cells and their microenvironment.


Cell Adhesion & Migration | 2013

Shape in migration: quantitative image analysis of migrating chemoresistant HCT-8 colon cancer cells.

Alessia Pasqualato; Vittorio Lei; Alessandra Cucina; Simona Dinicola; Fabrizio D'Anselmi; Sara Proietti; Maria Grazia Masiello; Alessandro Palombo; Mariano Bizzarri

Unsuccessful cytotoxic anticancer treatments may contribute to tumor morphologic instability and consequent tissue invasion, promoting the selection of a more malignant phenotype. Indeed, morphological changes have been demonstrated to be more pronounced in strongly vs. weakly metastatic cells. By means of normalized bending energy, we have previously quantitatively defined the link between cell shape modifications and the acquisition of a more malignant phenotype by 5-FU-resistant colon cancer cells (HCT-8FUres). Such changes were significantly correlated with an increase in motility speed. Herein, we propose a method to quantitatively analyze the shape of wild and chemoresistant HCT-8 migration front cells during wound healing assay. We evaluated the reliability of parameters (area/perimeter ratio [A/p], circularity, roundness, fractal dimension, and solidity) in describing the biological behavior of the two cell lines, enabling hence in distinguishing the chemoresistant line from the other one. We found solidity index the parameter that better described the difference between chemoresistant and wild cells. Moreover, solidity is able to capture the differences between chemoresistant and wild cells at each time point of the migration process. Indeed, motility speed was found to be inversely correlated with solidity, a quantitative index of cell deformability. Deformability is an outstanding hallmark of the process leading to metastatic spread; consequently, solidity may be considered a marker of acquired metastatic property.


Experimental Cell Research | 2016

Inositol induces mesenchymal-epithelial reversion in breast cancer cells through cytoskeleton rearrangement

Simona Dinicola; Gianmarco Fabrizi; Maria Grazia Masiello; Sara Proietti; Alessandro Palombo; Mirko Minini; Abdel Halim Harrath; Saleh H. Alwasel; Giulia Ricci; Angela Catizone; Alessandra Cucina; Mariano Bizzarri

Inositol displays multi-targeted effects on many biochemical pathways involved in epithelial-mesenchymal transition (EMT). As Akt activation is inhibited by inositol, we investigated if such effect could hamper EMT in MDA-MB-231 breast cancer cells. In cancer cells treated with pharmacological doses of inositol E-cadherin was increased, β-catenin was redistributed behind cell membrane, and metalloproteinase-9 was significantly reduced, while motility and invading capacity were severely inhibited. Those changes were associated with a significant down-regulation of PI3K/Akt activity, leading to a decrease in downstream signaling effectors: NF-kB, COX-2, and SNAI1. Inositol-mediated inhibition of PS1 leads to lowered Notch 1 release, thus contributing in decreasing SNAI1 levels. Overall, these data indicated that inositol inhibits the principal molecular pathway supporting EMT. Similar results were obtained in ZR-75, a highly metastatic breast cancer line. These findings are coupled with significant changes on cytoskeleton. Inositol slowed-down vimentin expression in cells placed behind the wound-healing edge and stabilized cortical F-actin. Moreover, lamellipodia and filopodia, two specific membrane extensions enabling cell migration and invasiveness, were no longer detectable after inositol addiction. Additionally, fascin and cofilin, two mandatory required components for F-actin assembling within cell protrusions, were highly reduced. These data suggest that inositol may induce an EMT reversion in breast cancer cells, suppressing motility and invasiveness through cytoskeleton modifications.


Toxicology in Vitro | 2015

Multiwalled carbon nanotube buckypaper induces cell cycle arrest and apoptosis in human leukemia cell lines through modulation of AKT and MAPK signaling pathways

Simona Dinicola; Maria Grazia Masiello; Sara Proietti; Pierpaolo Coluccia; Gianmarco Fabrizi; Alessandro Palombo; F. Micciulla; Silvia Bistarelli; Giulia Ricci; Angela Catizone; Giorgio De Toma; Mariano Bizzarri; S. Bellucci; Alessandra Cucina

MWCNT buckypaper (BP) shows physico-chemical and mechanical properties that make it potentially useful as a substrate in nano-bio interface research including in tissue engineering. When used as a scaffold material, BP comes into contact with host cells and surrounding tissues; therefore it is critical to determine its biocompatibility and interaction with living systems. The aim of this study was to investigate BP effects on cell growth, apoptosis and reactive oxygen species (ROS) production in three human leukemia cell lines HL-60, U-937 and K-562. BP was able to induce both the reduction of cell proliferation, associated with an arrest in G0/G1 phase of cell cycle and the increase of apoptosis in leukemic cell lines, thus exerting both cytostatic and cytotoxic effects. The growth inhibitory effect was likely mediated by the decrease of cyclins D, E, A, B1 levels and CDK4 expression; meanwhile, the apoptotic effect, not mediated by ROS production, was presumably due to the combined action of the survival and pro-apoptotic AKT and MAPK signal transduction pathways. These results raised the issue of biocompatibility of MWCNT BP for the creation of carbon nanotubes based scaffolds to utilize as prostheses in tissue engineering.


Life Sciences | 2016

Paradoxical E-cadherin increase in 5FU-resistant colon cancer is unaffected during mesenchymal-epithelial reversion induced by γ-secretase inhibition

Simona Dinicola; Alessia Pasqualato; Sara Proietti; Maria Grazia Masiello; Alessandro Palombo; Pierpaolo Coluccia; Rita Canipari; Angela Catizone; Giulia Ricci; Abdel Halim Harrath; Saleh H. Alwasel; Alessandra Cucina; Mariano Bizzarri

AIM Presenilin-1 (PS1), the main component of γ-secretase activity support a key role during Epithelial-Mesenchymal Transition (EMT) and chemoresistance acquisition by triggering a complex sequence of molecular events, including E-cadherin down-regulation. However, we hypothesize that EMT and chemoresistance should be deemed separate processes in HCT-8 colon cancer cells. MAIN METHODS HCT-8 and HCT-8FUres invasion was evaluated by trans-well assay. uPA activity was detected by zymography. Prostaglandin E2 levels were quantified using an ELISA kit. E-cadherin FL and CTF2, PS1, Notch1, Cyclin D1, COX2, SNAI1 and α-SMA expression were determined using Western blot technique. β-Catenin localization was observed by confocal microscopy. Cell apoptosis was evaluated by cytofluorimetric assay, and measurement of caspase-3 and cl-PARP. γ-Secretase activity was inhibited by DAPT, a γ-secretase inhibitor. KEY FINDINGS Chemoresistant HCT-8 underwent EMT that can be efficiently reversed by inhibiting PS1 activity, leading thus to a normalization of mostly of the pivotal features showed by the invasive cancer phenotype. Indeed, we observed decreased SNAI1 and Notch 1 activation, altogether with reduced E-cadherin cleavage. Concomitantly, resistant HCT-8 invasiveness was almost completely abolished. However, such reversion was not followed by any increase in apoptotic rate, not by changes in E-cadherin levels. Indeed, despite HCT-8FUres underwent an undeniable EMT, full-length E-cadherin levels were found remarkably higher than those observed in wild HCT-8. SIGNIFICANCE High E-cadherin concentration in presence of enhanced γ-secretase activity is incontestably a paradoxically result, highlighting that E-cadherin loss is not a pre-requisite for EMT. Additionally, EMT and chemoresistance acquisition in HCT-8 should be considered as distinct processes.

Collaboration


Dive into the Maria Grazia Masiello's collaboration.

Top Co-Authors

Avatar

Alessandra Cucina

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Mariano Bizzarri

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Sara Proietti

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Simona Dinicola

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Alessandro Palombo

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Angela Catizone

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Giulia Ricci

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Pierpaolo Coluccia

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gianmarco Fabrizi

Sapienza University of Rome

View shared research outputs
Researchain Logo
Decentralizing Knowledge