Network


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

Hotspot


Dive into the research topics where Carmen Mecca is active.

Publication


Featured researches published by Carmen Mecca.


Frontiers in Molecular Neuroscience | 2017

The Pathophysiological Role of Microglia in Dynamic Surveillance, Phagocytosis and Structural Remodeling of the Developing CNS

Cataldo Arcuri; Carmen Mecca; Roberta Bianchi; Ileana Giambanco; Rosario Donato

In vertebrates, during an early wave of hematopoiesis in the yolk sac between embryonic day E7.0 and E9.0, cells of mesodermal leaflet addressed to macrophage lineage enter in developing central nervous system (CNS) and originate the developing native microglial cells. Depending on the species, microglial cells represent 5–20% of glial cells resident in adult brain. Here, we briefly discuss some canonical functions of the microglia, i.e., cytokine secretion and functional transition from M1 to M2 phenotype. In addition, we review studies on the non-canonical functions of microglia such as regulation of phagocytosis, synaptic pruning, and sculpting postnatal neural circuits. In this latter context the contribution of microglia to some neurodevelopmental disorders is now well established. Nasu-Hakola (NHD) disease is considered a primary microgliopathy with alterations of the DNAX activation protein 12 (DAP12)-Triggering receptor expressed on myeloid cells 2 (TREM-2) signaling and removal of macromolecules and apoptotic cells followed by secondary microglia activation. In Rett syndrome Mecp2-/- microglia shows a substantial impairment of phagocytic ability, although the role of microglia is not yet clear. In a mouse model of Tourette syndrome (TS), microglia abnormalities have also been described, and deficient microglia-mediated neuroprotection is obvious. Here we review the role of microglial cells in neurodevelopmental disorders without inflammation and on the complex role of microglia in developing CNS.


International Journal of Molecular Sciences | 2018

Microglia and Aging: The Role of the TREM2–DAP12 and CX3CL1-CX3CR1 Axes

Carmen Mecca; Ileana Giambanco; Rosario Donato; Cataldo Arcuri

Depending on the species, microglial cells represent 5–20% of glial cells in the adult brain. As the innate immune effector of the brain, microglia are involved in several functions: regulation of inflammation, synaptic connectivity, programmed cell death, wiring and circuitry formation, phagocytosis of cell debris, and synaptic pruning and sculpting of postnatal neural circuits. Moreover, microglia contribute to some neurodevelopmental disorders such as Nasu-Hakola disease (NHD), and to aged-associated neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and others. There is evidence that human and rodent microglia may become senescent. This event determines alterations in the microglia activation status, associated with a chronic inflammation phenotype and with the loss of neuroprotective functions that lead to a greater susceptibility to the neurodegenerative diseases of aging. In the central nervous system (CNS), Triggering Receptor Expressed on Myeloid Cells 2-DNAX activation protein 12 (TREM2-DAP12) is a signaling complex expressed exclusively in microglia. As a microglial surface receptor, TREM2 interacts with DAP12 to initiate signal transduction pathways that promote microglial cell activation, phagocytosis, and microglial cell survival. Defective TREM2-DAP12 functions play a central role in the pathogenesis of several diseases. The CX3CL1 (fractalkine)-CX3CR1 signaling represents the most important communication channel between neurons and microglia. The expression of CX3CL1 in neurons and of its receptor CX3CR1 in microglia determines a specific interaction, playing fundamental roles in the regulation of the maturation and function of these cells. Here, we review the role of the TREM2-DAP12 and CX3CL1-CX3CR1 axes in aged microglia and the involvement of these pathways in physiological CNS aging and in age-associated neurodegenerative diseases.


Ecancermedicalscience | 2015

miRNAs and resistance to EGFR-TKIs in EGFR-mutant non-small cell lung cancer: beyond 'traditional mechanisms' of resistance.

Biagio Ricciuti; Carmen Mecca; Matteo Cenci; Giulia Costanza Leonardi; Lorenzo Perrone; Clelia Mencaroni; Lucio Crinò; Francesco Grignani; Sara Baglivo; Rita Chiari; Angelo Sidoni; Luca Paglialunga; Maria Francesca Currà; Emanuele Murano; V. Minotti; Giulio Metro

Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) have dramatically changed the prognosis of advanced non-small cell lung cancers (NSCLCs) that harbour specific EGFR activating mutations. However, the efficacy of an EGFR-TKI is limited by the onset of acquired resistance, usually within one year, in virtually all treated patients. Moreover, a small percentage of EGFR-mutant NSCLCs do not respond to an EGFR-TKI, thus displaying primary resistance. At the present time, several mechanisms of either primary and acquired resistance have been elucidated, and new drugs are currently under preclinical and clinical development in order to overcome resistance to treatment. Nevertheless, there still remains much to be thoroughly investigated, as so far research has mainly focused on the role of proteincoding genes involved in resistance to EGFR-TKIs. On the other hand, in line with the data underscoring the relevance of non-coding RNAs in the pathogenesis of lung cancer and modulation of response to systemic therapies, microRNAs (miRNAs) have been supposed to play an important role in resistance to EGFR-TKIs. The aim of this review is to briefly summarise the existing relationship between miRNAs and resistance to EGFR-TKIs, and also focusing on the possible clinical applications of miRNAs in reverting and overcoming such resistance.


Frontiers in Cellular Neuroscience | 2018

PP242 Counteracts Glioblastoma Cell Proliferation, Migration, Invasiveness and Stemness Properties by Inhibiting mTORC2/AKT

Carmen Mecca; Ileana Giambanco; Stefano Bruscoli; Oxana Bereshchenko; Bernard Fioretti; Carlo Riccardi; Rosario Donato; Cataldo Arcuri

Glioblastoma multiforme (GBM) is the most malignant brain tumor and is associated with poor prognosis due to its thorny localization, lack of efficacious therapies and complex biology. Among the numerous pathways driving GBM biology studied so far, PTEN/phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT/mechanistic target of rapamycin (mTOR) signaling plays a pivotal role, as it controls cell survival, proliferation and metabolism and is involved in stem cell maintenance. In front of recent and numerous evidences highlighting mTOR upregulation in GBM, all the strategies developed to inhibit this pathway have been substantially unsuccessful. Our study focused on mTOR complex 2 (mTORC2) to understand its involvement in GBM cell growth, proliferation, migration and invasiveness. We utilized an in vitro model, characterized by various genetic alterations (i.e., GL15, U257, U87MG and U118MG cell lines) in order to achieve the clonal heterogeneity observed in vivo. Additionally, being the U87MG cell line endowed with glioblastoma stem cells (GSCs), we also investigated the role of the PTEN/PI3K/AKT/mTOR pathway in this specific cell population, which is responsible for GBM relapse. We provide further insights that explain the reasons for the failure of numerous clinical trials conducted to date targeting PI3K or mTOR complex 1 (mTORC1) with rapamycin and its analogs. Additionally, we show that mTORC2 might represent a potential clinically valuable target for GBM treatment, as proliferation, migration and GSC maintenance appear to be mTORC2-dependent. In this context, we demonstrate that the novel ATP-competitive mTOR inhibitor PP242 effectively targets both mTORC1 and mTORC2 activation and counteracts cell proliferation via the induction of high autophagy levels, besides reducing cell migration, invasiveness and stemness properties.


Mediators of Inflammation | 2017

Neutral Sphingomyelinase Behaviour in Hippocampus Neuroinflammation of MPTP-Induced Mouse Model of Parkinson’s Disease and in Embryonic Hippocampal Cells

Samuela Cataldi; Cataldo Arcuri; Stéphane Hunot; François-Pierre Légeron; Carmen Mecca; Mercedes Garcia-Gil; Andrea Lazzarini; Michela Codini; Tommaso Beccari; Anna Tasegian; Bernard Fioretti; Giovanna Traina; Francesco Saverio Ambesi-Impiombato; Francesco Curcio; Elisabetta Albi

Neutral sphingomyelinase is known to be implicated in growth arrest, differentiation, proliferation, and apoptosis. Although previous studies have reported the involvement of neutral sphingomyelinase in hippocampus physiopathology, its behavior in the hippocampus during Parkinsons disease remains undetected. In this study, we show an upregulation of inducible nitric oxide synthase and a downregulation of neutral sphingomyelinase in the hippocampus of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine- (MPTP-) induced mouse model of Parkinsons disease. Moreover, the stimulation of neutral sphingomyelinase activity with vitamin 1,25-dihydroxyvitamin D3 reduces specifically saturated fatty acid sphingomyelin by making sphingomyelin a less rigid molecule that might influence neurite plasticity. The possible biological relevance of the increase of neutral sphingomyelinase in Parkinsons disease is discussed.


Nanomaterials | 2018

Probing Internalization Effects and Biocompatibility of Ultrasmall Zirconium Metal-Organic Frameworks UiO-66 NP in U251 Glioblastoma Cancer Cells

Cataldo Arcuri; Lorenzo Monarca; Francesco Ragonese; Carmen Mecca; Stefano Bruscoli; Stefano Giovagnoli; Rosario Donato; Oxana Bereshchenko; Bernard Fioretti; Ferdinando Costantino

The synthesis of ultrasmall UiO-66 nanoparticles (NPs) with an average size of 25 nm, determined by X-ray powder diffraction and electron microscopies analysis, is reported. The NPs were stabilized in water by dialyzing the NP from the DMF used for the synthesis. DLS measurements confirmed the presence of particles of 100 nm, which are spherical aggregates of smaller particles of 20–30 nm size. The NP have a BET surface area of 700 m2/g with an external surface area of 300 m2/g. UiO-66_N (UiO-66 nanoparticles) were loaded with acridine orange as fluorescent probe. UV-vis spectroscopy analysis revealed no acridine loss after 48 h of agitation in simulated body fluid. The biocompatibility of UiO-66_N was evaluated in human glioblastoma (GBM) cell line U251, the most malignant (IV grade of WHO classification) among brain tumors. In U251 cells, UiO-66_N are inert since they do not alter the cell cycle, the viability, migration properties, and the expression of kinases involved in cancer cell growth. The internalization process was evident after a few hours of incubation. After 24 h, UiO-66_N@Acr (UiO-66_N loaded with acridine orange) were detectable around the nuclei of the cells. These data suggest that small UiO-66 are biocompatible NP and could represent a potential carrier for drug delivery in glioblastoma therapies.


Frontiers in Cellular Neuroscience | 2018

Effect of Vitamin D in HN9.10e Embryonic Hippocampal Cells and in Hippocampus from MPTP-Induced Parkinson’s Disease Mouse Model

Samuela Cataldi; Cataldo Arcuri; Stéphane Hunot; Carmen Mecca; Michela Codini; Maria E. Laurenti; Ivana Ferri; Elisabetta Loreti; Mercedes Garcia-Gil; Giovanna Traina; Carmela Conte; Francesco Saverio Ambesi-Impiombato; Tommaso Beccari; Francesco Curcio; Elisabetta Albi

It has long been proven that neurogenesis continues in the adult brains of mammals in the dentatus gyrus of the hippocampus due to the presence of neural stem cells. Although a large number of studies have been carried out to highlight the localization of vitamin D receptor in hippocampus, the expression of vitamin D receptor in neurogenic dentatus gyrus of hippocampus in Parkinson’s disease (PD) and the molecular mechanisms triggered by vitamin D underlying the production of differentiated neurons from embryonic cells remain unknown. Thus, we performed a preclinical in vivo study by inducing PD in mice with MPTP and showed a reduction of glial fibrillary acidic protein (GFAP) and vitamin D receptor in the dentatus gyrus of hippocampus. Then, we performed an in vitro study by inducing embryonic hippocampal cell differentiation with vitamin D. Interestingly, vitamin D stimulates the expression of its receptor. Vitamin D receptor is a transcription factor that probably is responsible for the upregulation of microtubule associated protein 2 and neurofilament heavy polypeptide genes. The latter increases heavy neurofilament protein expression, essential for neurofilament growth. Notably N-cadherin, implicated in activity for dendritic outgrowth, is upregulated by vitamin D.


Italian journal of anatomy and embryology | 2017

An in vitro study of the mTORC1/2 inhibitor PP242 in glioblastoma multiforme

Carmen Mecca; Ileana Giambanco; Rosario Donato; Cataldo Arcuri

mTOR is a kinase complex involved in cell growth, proliferation, survival, metabolism and migration. The aberrant activation of mTOR has been previously demonstrated in glioblastoma multiforme (GBM), making it an interesting target for therapeutic approaches [1]. Unfortunately, the attempts to block mTOR activity made so far had disappointing clinical efficacy, as the mTOR inhibitor Rapamycin and analogs only target mTOR complex 1 (mTORC1) while mTOR actually exists in two distinct complexes, namely mTORC1 and mTOR complex 2 (mTORC2) that differ in terms of both regulation mechanisms and functions [2,3]. mTORC1 is inhibited by Rapamycin and acts as a downstream effector of the PTEN/PI3K/Akt pathway, linking growth factors, amino acids, ATP and O2 signals to protein translation, cell growth, proliferation and survival. Differently, mTORC2 is insensible to Rapamycin and acts as an upstream activator of Akt via phosphorylation of serine 473 [3]. To analyze the contribution of mTORC1/2 to GBM biology, we studied the in vitro effect of PP242, a novel mTORC1/2 inhibitor, on glioma cell lines of different malignancy degree, and compared it to the effect of Rapamycin and of the irreversible PI3K inhibitor, Wortmannin. Our results suggest that the inhibition of both mTOR complexes with PP242 induces sustained levels of autophagy that causes G0/G1 cell cycle arrest and a significantly reduction of cell viability, proliferation and migration. Additionally, we observed that administration of PP242 in U87MG cell line prevents stem cell growth, which results in the inhibition of neurospheres formation. This data confirms the pivotal role of mTOR in glioblastoma cells biology and expand upon this evidence suggesting a prominent role of the mTOR complex 2 in glioblastoma cell growth, migration and survival, and indicate that the mTORC2 might represent clinically valuable target in GMB.


ONCOSCIENCE | 2014

Non-coding RNAs in lung cancer

Biagio Ricciuti; Carmen Mecca; Lucio Crinò; Sara Baglivo; Matteo Cenci; Giulio Metro


Frontiers in Bioscience | 2017

Microglia-glioma cross-talk: a two way approach to new strategies against glioma.

Cataldo Arcuri; Bernard Fioretti; Roberta Bianchi; Carmen Mecca; Claudia Tubaro; Tommaso Beccari; Fabio Franciolini; Ileana Giambanco; Rosario Donato

Collaboration


Dive into the Carmen Mecca's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge