Network


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

Hotspot


Dive into the research topics where Antonietta Arcella is active.

Publication


Featured researches published by Antonietta Arcella.


Journal of Neurochemistry | 2007

Capsaicin-induced apoptosis of glioma cells is mediated by TRPV1 vanilloid receptor and requires p38 MAPK activation.

Consuelo Amantini; Michela Mosca; Massimo Nabissi; Roberta Lucciarini; Stefano Caprodossi; Antonietta Arcella; Felice Giangaspero; Giorgio Santoni

We provide evidence on the expression of the transient receptor potential vanilloid type‐1 (TRPV1) by glioma cells, and its involvement in capsaicin (CPS)‐induced apoptosis. TRPV1 mRNA was identified by quantitative RT‐PCR in U373, U87, FC1 and FLS glioma cells, with U373 cells showing higher, and U87, FC1 and FLS cells lower TRPV1 expression as compared with normal human astrocytes. By flow cytometry we found that a substantial portion of both normal human astrocytes, and U87 and U373 glioma cells express TRPV1 protein. Moreover, we analyzed the expression of TRPV1 at mRNA and protein levels of glioma tissues with different grades. We found that TRPV1 gene and protein expression inversely correlated with glioma grading, with marked loss of TRPV1 expression in the majority of grade IV glioblastoma multiforme. We also described that CPS trigger apoptosis of U373, but not U87 cells. CPS‐induced apoptosis involved Ca2+ influx, p38 but not extracellular signal‐regulated mitogen‐activated protein kinase activation, phosphatidylserine exposure, mitochondrial permeability transmembrane pore opening and mitochondrial transmembrane potential dissipation, caspase 3 activation and oligonucleosomal DNA fragmentation. TRPV1 was functionally implicated in these events as they were markedly inhibited by the TRPV1 antagonist, capsazepine. Finally, p38 but not extracellular signal‐regulated protein kinase activation was required for TRPV1‐mediated CPS‐induced apoptosis of glioma cells.


Endocrine-related Cancer | 2009

Expression of aryl hydrocarbon receptor (AHR) and AHR-interacting protein in pituitary adenomas: pathological and clinical implications

Marie Lise Jaffrain-Rea; M. Angelini; Donatella Gargano; Maria A. Tichomirowa; Adrian Daly; Jean François Vanbellinghen; Emanuela D'Innocenzo; Anne Barlier; Felice Giangaspero; Vincenzo Esposito; L. Ventura; Antonietta Arcella; Marily Theodoropoulou; Luciana A. Naves; Carmen Fajardo; Sabina Zacharieva; V. Rohmer; Thierry Brue; Alberto Gulino; Giampaolo Cantore; Edoardo Alesse; Albert Beckers

Germline mutations of the aryl hydrocarbon receptor (AHR)-interacting protein (AIP) gene confer a predisposition to pituitary adenomas (PA), usually in the setting of familial isolated PA. To provide further insights into the possible role of AIP in pituitary tumour pathogenesis, the expression of AIP and AHR was determined by real-time RT-PCR and/or immunohistochemistry (IHC) in a large series of PA (n=103), including 17 with AIP mutations (AIP(mut)). Variable levels of AIP and AHR transcripts were detected in all PA, with a low AHR expression (P<0.0001 versus AIP). Cytoplasmic AIP and AHR were detected by IHC in 84.0 and 38.6% of PA respectively, and significantly correlated with each other (P=0.006). Nuclear AHR was detected in a minority of PA (19.7%). The highest AIP expression was observed in somatotrophinomas and non-secreting (NS) PA, and multivariate analysis in somatotrophinomas showed a significantly lower AIP immunostaining in invasive versus non-invasive cases (P=0.019). AIP expression was commonly low in other secreting PA. AIP immunostaining was abolished in a minority of AIP(mut) PA, with a frequent loss of cytoplasmic AHR and no evidence of nuclear AHR. In contrast, AIP overexpression in a subset of NS PA could be accompanied by nuclear AHR immunopositivity. We conclude that down-regulation of AIP and AHR may be involved in the aggressiveness of somatotrophinomas. Overall, IHC is a poorly sensitive tool for the screening of AIP mutations. Data obtained on AHR expression suggest that AHR signalling may be differentially affected according to PA phenotype.


Neuro-oncology | 2005

Pharmacological blockade of group II metabotropic glutamate receptors reduces the growth of glioma cells in vivo

Antonietta Arcella; Giulia Carpinelli; Giuseppe Battaglia; Mara D'Onofrio; Filippo Santoro; Richard Teke Ngomba; Valeria Bruno; Paola Casolini; Felice Giangaspero; Ferdinando Nicoletti

U87MG human glioma cells in cultures expressed metabotropic glutamate (mGlu) receptors mGlu2 and mGlu3. Addition of the mGlu2/3 receptor antagonist LY341495 to the cultures reduced cell growth, expression of cyclin D1/2, and activation of the MAP kinase and phosphatidylinositol-3-kinase pathways. This is in line with the evidence that activation of mGlu2/3 receptors sustains glioma cell proliferation. U87MG cells were either implanted under the skin (1x10(6) cells/0.5 ml) or infused into the caudate nucleus (0.5x10(6) cells/5 microl) of nude mice. Animals were treated for 28 days with mGlu receptor antagonists by means of subcutaneous osmotic minipumps. Treatments with LY341495 or (2S)-alpha-ethylglutamate (both infused at a rate of 1 mg/kg per day) reduced the size of tumors growing under the skin. Infusion of LY341495 (10 mg/kg per day) also reduced the growth of brain tumors, as assessed by magnetic resonance imaging analysis carried out every seven days. The effect of drug treatment was particularly evident during the exponential phase of tumor growth, that is, between the third and the fourth week following cell implantation. Immunohistochemical analysis showed that U87MG cells retained the expression of mGlu2/3 receptors when implanted into the brain of nude mice. These data suggest that mGlu2/3 receptor antagonists are of potential use in the experimental treatment of malignant gliomas.


Journal of Neurochemistry | 2003

Pharmacological blockade of mGlu2/3 metabotropic glutamate receptors reduces cell proliferation in cultured human glioma cells

Mara D'Onofrio; Antonietta Arcella; Valeria Bruno; Richard Teke Ngomba; Giuseppe Battaglia; Vincenza Lombari; Giuseppe Ragona; Antonella Calogero; Ferdinando Nicoletti

Glial cell proliferation in culture is under the control of metabotropic glutamate (mGlu) receptors. We have examined whether this control extends to human glioma cells. Primary cultures were prepared from surgically removed human glioblastomas. RT‐PCR combined with western blot analysis showed that most of the cultures (eight out of 11) expressed group‐II mGlu receptors. In two selected cultures (MZC‐12 and FCN‐9), the mGlu2/3 receptor antagonist, LY341495, slowed cell proliferation when applied to the growth medium from the second day after plating. This effect was reversible because linear cell growth was restored after washing out the drug. LY341495 reduced glioma cell proliferation at concentrations lower than 100 nm, which are considered as selective for mGlu2/3 receptors. In addition, its action was mimicked by the putative mGlu2/3 receptor antagonist (2S)‐α‐ethylglutamate. The anti‐proliferative effect of LY341495 was confirmed by measuring [methyl‐3H]‐thymidine incorporation in cultures arrested in G0 phase of the cell cycle and then stimulated to proliferate by the addition of 10% fetal calf serum or 100 ng/mL of epidermal growth factor (EGF). In cultures treated with EGF, LY341495 was also able to reduce the stimulation of the mitogen‐activated protein kinase (MAPK) pathway, as well as the induction of cyclin D1. Both effects, as well as decreased [methyl‐3H]‐thymidine incorporation, were partially reduced by co‐addition of the potent mGlu2/3 receptor agonist, LY379268. We conclude that activation of group‐II mGlu receptors supports the growth of human glioma cells in culture and that antagonists of these receptors should be tested for their ability to reduce tumour growth in vivo.


The Journal of Neuroscience | 2006

Pharmacological activation of mGlu4 metabotropic glutamate receptors inhibits the growth of medulloblastomas.

Luisa Iacovelli; Antonietta Arcella; Giuseppe Battaglia; Simonetta Pazzaglia; Eleonora Aronica; Paola Spinsanti; Alessandra Caruso; Enrico De Smaele; Anna Saran; Alberto Gulino; Mara D'Onofrio; Felice Giangaspero; Ferdinando Nicoletti

Moving from the evidence that activation of type 4 metabotropic glutamate (mGlu4) receptors inhibits proliferation and promotes differentiation of cerebellar granule cell neuroprogenitors, we examined the expression and function of mGlu4 receptors in medulloblastoma cells. mGlu4 receptors were expressed in 46 of 60 human medulloblastoma samples. Expression varied in relation to the histotype (nodular desmoplastic>classic≫large-cell anaplastic) and was inversely related to tumor severity, spreading, and recurrence. mGlu4 receptors were also found in D283med, D341med, and DAOY medulloblastoma cell lines, where receptor activation with the selective enhancer PHCCC inhibited adenylyl cyclase and the phosphatidylinositol-3-kinase pathway without affecting the mitogen-activated protein kinase, Sonic Hedgehog, and Wnt pathways. Interestingly, mGlu4 receptor activation reduced DNA synthesis and cell proliferation in all three cell lines. This effect was abrogated by the phosphatidylinositol-3-kinase inhibitor LY294002 [2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one]. In in vivo experiments, repeated subcutaneous injections of N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC) reduced the growth of D283med and DAOY cell xenografts in nude mice. More remarkably, subcutaneous or intracranial injections of PHCCC during the first week of life prevented the development of medulloblastomas in mice lacking one Patched-1 allele and x-irradiated 1 d after birth. These data suggest that mGlu4 receptor enhancers are promising drugs for the treatment of medulloblastomas.


Cancer Cell International | 2004

Inhibition of cell growth by EGR-1 in human primary cultures from malignant glioma.

Antonella Calogero; Vincenza Lombari; Giorgia De Gregorio; Antonio Porcellini; Severine Ucci; Antonietta Arcella; Riccardo Caruso; Franco Maria Gagliardi; Alberto Gulino; Gaetano Lanzetta; Luigi Frati; Dan Mercola; Giuseppe Ragona

BackgroundThe aim of this work was to investigate in vitro the putative role of EGR-1 in the growth of glioma cells. EGR-1 expression was examined during the early passages in vitro of 17 primary cell lines grown from 3 grade III and from 14 grade IV malignant astrocytoma explants. The explanted tumors were genetically characterized at the p53, MDM2 and INK4a/ARF loci, and fibronectin expression and growth characteristics were examined. A recombinant adenovirus overexpressing EGR-1 was tested in the primary cell lines.ResultsLow levels of EGR-1 protein were found in all primary cultures examined, with lower values present in grade IV tumors and in cultures carrying wild-type copies of p53 gene. The levels of EGR-1 protein were significantly correlated to the amount of intracellular fibronectin, but only in tumors carrying wild-type copies of the p53 gene (R = 0,78, p = 0.0082). Duplication time, plating efficiency, colony formation in agarose, and contact inhibition were also altered in the p53 mutated tumor cultures compared to those carrying wild-type p53. Growth arrest was achieved in both types of tumor within 1–2 weeks following infection with a recombinant adenovirus overexpressing EGR-1 but not with the control adenovirus.ConclusionsSuppression of EGR-1 is a common event in gliomas and in most cases this is achieved through down-regulation of gene expression. Expression of EGR-1 by recombinant adenovirus infection almost completely abolishes the growth of tumor cells in vitro, regardless of the mutational status of the p53 gene.


The Journal of Neuroscience | 2004

PHCCC, a specific enhancer of type 4 metabotropic glutamate receptors, reduces proliferation and promotes differentiation of cerebellar granule cell neuroprecursors

A. M. Canudas; V. Di Giorgi-Gerevini; Luisa Iacovelli; G. Nano; Mara D'Onofrio; Antonietta Arcella; Felice Giangaspero; Carla L. Busceti; L. Ricci-Vitiani; Giuseppe Battaglia; Ferdinando Nicoletti; Daniela Melchiorri

Exposure of immature rat cerebellar granule cell cultures to the type 4 metabotropic glutamate (mGlu4) receptor enhancer N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC) reduced [3H]thymidine incorporation. Its action was sensitive to the growth conditions and was attenuated by two mGlu4 receptor antagonists. An antiproliferative action of PHCCC was also seen in cultures from wild-type, but not mGlu4, knock-out mice. At least in rat cultures, PHCCC was not neurotoxic and enhanced neuritogenesis. Although PHCCC reduced the increase in cAMP formation and phospho-AKT levels induced by forskolin, none of these transduction pathways significantly contributed to the reduction of [3H]thymidine incorporation. Interestingly, PHCCC reduced the expression of Gli-1, a transcription factor that mediates the mitogenic action of Sonic hedgehog. Finally, we treated newborn rats with PHCCC either intracerebrally (infusion of 5 nmol/2 μl in the cerebellar region once every other day) or systemically (5 mg/kg, i.p., once daily) from postnatal days 3-9. Local infusion of PHCCC induced substantial changes in the morphology of the developing cerebellum. In contrast, systemic injection of PHCCC induced only morphological abnormalities of the cerebellar lobule V, which became visible 11 d after the end of the treatment. These data suggest that mGlu4 receptors are involved in the regulation of cerebellar development.


Nature Communications | 2013

Proteolysis of MOB1 by the ubiquitin ligase praja2 attenuates Hippo signalling and supports glioblastoma growth

Luca Lignitto; Antonietta Arcella; Maria Sepe; Laura Rinaldi; Rossella Delle Donne; Adriana Gallo; Eduard Stefan; Verena Bachmann; Maria Antonietta Oliva; Clelia Tiziana Storlazzi; Alberto L'Abbate; Arturo Brunetti; Sara Gargiulo; Matteo Gramanzini; Luigi Insabato; Corrado Garbi; Max E. Gottesman; Antonio Feliciello

Human glioblastoma is the most frequent and aggressive form of brain tumour in the adult population. Proteolytic turnover of tumour suppressors by the ubiquitin–proteasome system is a mechanism that tumour cells can adopt to sustain their growth and invasiveness. However, the identity of ubiquitin–proteasome targets and regulators in glioblastoma are still unknown. Here we report that the RING ligase praja2 ubiquitylates and degrades Mob, a core component of NDR/LATS kinase and a positive regulator of the tumour-suppressor Hippo cascade. Degradation of Mob through the ubiquitin–proteasome system attenuates the Hippo cascade and sustains glioblastoma growth in vivo. Accordingly, accumulation of praja2 during the transition from low- to high-grade glioma is associated with significant downregulation of the Hippo pathway. These findings identify praja2 as a novel upstream regulator of the Hippo cascade, linking the ubiquitin proteasome system to deregulated glioblastoma growth.


Neuroscience | 2010

Tissue plasminogen activator and urokinase plasminogen activator in human epileptogenic pathologies

Anand M. Iyer; Emanuele Zurolo; Karin Boer; Johannes C. Baayen; Felice Giangaspero; Antonietta Arcella; G. Di Gennaro; Vincenzo Esposito; Wim G. M. Spliet; P.C. van Rijen; Dirk Troost; Jan A. Gorter; Eleonora Aronica

A growing body of evidence demonstrates the involvement of plasminogen activators (PAs) in a number of physiologic and pathologic events in the CNS. Induction of both tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA) has been observed in different experimental models of epilepsy and tPA has been implicated in the mechanisms underlying seizure activity. We investigated the expression and the cellular distribution of tPA and uPA in several epileptogenic pathologies, including hippocampal sclerosis (HS; n=6), and developmental glioneuronal lesions, such as focal cortical dysplasia (FCD, n=6), cortical tubers in patients with the tuberous sclerosis complex (TSC; n=6) and in gangliogliomas (GG; n=6), using immuno-cytochemical, western blot and real-time quantitative PCR analysis. TPA and uPA immunostaining showed increased expression within the epileptogenic lesions compared to control specimens in both glial and neuronal cells (hippocampal neurons in HS and dysplastic neurons in FCD, TSC and GG specimens). Confocal laser scanning microscopy confirmed expression of both proteins in astrocytes and microglia, as well as in microvascular endothelium. Immunoblot demonstrated also up-regulation of the uPA receptor (uPAR; P<0.05). Increased expression of tPA, uPA, uPAR and tissue PA inhibitor type mRNA levels was also detected by PCR analysis in different epileptogenic pathologies (P<0.05). Our data support the role of PA system components in different human focal epileptogenic pathologies, which may critically influence neuronal activity, inflammatory response, as well as contributing to the complex remodeling of the neuronal networks occurring in epileptogenic lesions.


Brain Pathology | 2012

KIAA1549-BRAF Fusions and IDH Mutations Can Coexist in Diffuse Gliomas of Adults

Manuela Badiali; Vincent Gleize; Sophie Paris; Loredana Moi; Selma Elhouadani; Antonietta Arcella; Roberta Morace; Manila Antonelli; Francesca R. Buttarelli; Dominique Figarella Branger; Young-Ho Kim; Hiroko Ohgaki; Karima Mokhtari; Marc Sanson; Felice Giangaspero

KIAA1549‐BRAF fusion gene and isocitrate dehydrogenase (IDH) mutations are considered two mutually exclusive genetic events in pilocytic astrocytomas and diffuse gliomas, respectively. We investigated the presence of the KIAA1549‐BRAF fusion gene in conjunction with IDH mutations and 1p/19q loss in 185 adult diffuse gliomas. Moreover BRAFv600E mutation was also screened. The KIAA1549‐BRAF fusion gene was evaluated by reverse‐transcription polymerase chain reaction (RT‐PCR) and sequencing. We found IDH mutations in 125 out 175 cases (71.4%). There were KIAA1549‐BRAF fusion gene in 17 out of 180 (9.4%) cases and BRAFv600E in 2 out of 133 (1.5%) cases. In 11 of these 17 cases, both IDH mutations and the KIAA1549‐BRAF fusion were present, as independent molecular events. Moreover, 6 of 17 cases showed co‐presence of 1p/19q loss, IDH mutations and KIAA1549‐BRAF fusion. Among the 17 cases with KIAA1549‐BRAF fusion gene 15 (88.2%) were oligodendroglial neoplasms. Similarly, the two cases with BRAFv600E mutation were both oligodendroglioma and one had IDH mutations and 1p/19q co‐deletion. Our results suggest that in a small fraction of diffuse gliomas, KIAA1549‐BRAF fusion gene and BRAFv600E mutation may be responsible for deregulation of the Ras‐RAF‐ERK signaling pathway. Such alterations are more frequent in oligodendroglial neoplasm and may be co‐present with IDH mutations and 1p/19q loss.

Collaboration


Dive into the Antonietta Arcella's collaboration.

Top Co-Authors

Avatar

Felice Giangaspero

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Vincenzo Esposito

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Valeria Bruno

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Manila Antonelli

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Maura Massimino

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge