Network


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

Hotspot


Dive into the research topics where Fabio Franciolini is active.

Publication


Featured researches published by Fabio Franciolini.


American Journal of Physiology-cell Physiology | 2010

CXCL12-induced glioblastoma cell migration requires intermediate conductance Ca2+-activated K+ channel activity

Miriam Sciaccaluga; Bernard Fioretti; Luigi Catacuzzeno; Francesca Pagani; Cristina Bertollini; Maria Rosito; Myriam Catalano; Giuseppina D'Alessandro; Antonio Santoro; Giampaolo Cantore; Davide Ragozzino; Emilia Castigli; Fabio Franciolini; Cristina Limatola

The activation of ion channels is crucial during cell movement, including glioblastoma cell invasion in the brain parenchyma. In this context, we describe for the first time the contribution of intermediate conductance Ca(2+)-activated K (IK(Ca)) channel activity in the chemotactic response of human glioblastoma cell lines, primary cultures, and freshly dissociated tissues to CXC chemokine ligand 12 (CXCL12), a chemokine whose expression in glioblastoma has been correlated with its invasive capacity. We show that blockade of the IK(Ca) channel with its specific inhibitor 1-[(2-chlorophenyl) diphenylmethyl]-1H-pyrazole (TRAM-34) or IK(Ca) channel silencing by short hairpin RNA (shRNA) completely abolished CXCL12-induced cell migration. We further demonstrate that this is not a general mechanism in glioblastoma cell migration since epidermal growth factor (EGF), which also activates IK(Ca) channels in the glioblastoma-derived cell line GL15, stimulate cell chemotaxis even if the IK(Ca) channels have been blocked or silenced. Furthermore, we demonstrate that both CXCL12 and EGF induce Ca(2+) mobilization and IK(Ca) channel activation but only CXCL12 induces a long-term upregulation of the IK(Ca) channel activity. Furthermore, the Ca(2+)-chelating agent BAPTA-AM abolished the CXCL12-induced, but not the EGF-induced, glioblastoma cell chemotaxis. In addition, we demonstrate that the extracellular signal-regulated kinase (ERK)1/2 pathway is only partially implicated in the modulation of CXCL12-induced glioblastoma cell movement, whereas the phosphoinositol-3 kinase (PI3K) pathway is not involved. In contrast, EGF-induced glioblastoma migration requires both ERK1/2 and PI3K activity. All together these findings suggest that the efficacy of glioblastoma invasiveness might be related to an array of nonoverlapping mechanisms activated by different chemotactic agents.


Cell Death and Disease | 2013

KCa3.1 channels are involved in the infiltrative behavior of glioblastoma in vivo

Giuseppina D'Alessandro; Myriam Catalano; Miriam Sciaccaluga; Giuseppina Chece; R. Cipriani; Maria Rosito; Alfonso Grimaldi; Clotilde Lauro; G. Cantore; Antonio Santoro; Bernard Fioretti; Fabio Franciolini; Heike Wulff; Cristina Limatola

Glioblastoma multiforme (GBM) is a diffuse brain tumor characterized by high infiltration in the brain parenchyma rendering the tumor difficult to eradicate by neurosurgery. Efforts to identify molecular targets involved in the invasive behavior of GBM suggested ion channel inhibition as a promising therapeutic approach. To determine if the Ca2+-dependent K+ channel KCa3.1 could represent a key element for GBM brain infiltration, human GL-15 cells were xenografted into the brain of SCID mice that were then treated with the specific KCa3.1 blocker TRAM-34 (1-((2-chlorophenyl) (diphenyl)methyl)-1H-pyrazole). After 5 weeks of treatment, immunofluorescence analyses of cerebral slices revealed reduced tumor infiltration and astrogliosis surrounding the tumor, compared with untreated mice. Significant reduction of tumor infiltration was also observed in the brain of mice transplanted with KCa3.1-silenced GL-15 cells, indicating a direct effect of TRAM-34 on GBM-expressed KCa3.1 channels. As KCa3.1 channels are also expressed on microglia, we investigated the effects of TRAM-34 on microglia activation in GL-15 transplanted mice and found a reduction of CD68 staining in treated mice. Similar results were observed in vitro where TRAM-34 reduced both phagocytosis and chemotactic activity of primary microglia exposed to GBM-conditioned medium. Taken together, these results indicate that KCa3.1 activity has an important role in GBM invasiveness in vivo and that its inhibition directly affects glioma cell migration and reduces astrocytosis and microglia activation in response to tumor-released factors. KCa3.1 channel inhibition therefore constitutes a potential novel therapeutic approach to reduce GBM spreading into the surrounding tissue.


Frontiers in Cellular Neuroscience | 2013

K + channelepsy: progress in the neurobiology of potassium channels and epilepsy

Maria Cristina D'Adamo; Luigi Catacuzzeno; Giuseppe Di Giovanni; Fabio Franciolini; Mauro Pessia

K+ channels are important determinants of seizure susceptibility. These membrane proteins, encoded by more than 70 genes, make the largest group of ion channels that fine-tune the electrical activity of neuronal and non-neuronal cells in the brain. Their ubiquity and extremely high genetic and functional diversity, unmatched by any other ion channel type, place K+ channels as primary targets of genetic variations or perturbations in K+-dependent homeostasis, even in the absence of a primary channel defect. It is therefore not surprising that numerous inherited or acquired K+ channels dysfunctions have been associated with several neurologic syndromes, including epilepsy, which often generate confusion in the classification of the associated diseases. Therefore, we propose to name the K+ channels defects underlying distinct epilepsies as “K+ channelepsies,” and introduce a new nomenclature (e.g., Kx.y-channelepsy), following the widely used K+ channel classification, which could be also adopted to easily identify other channelopathies involving Na+ (e.g., Navx.y-phenotype), Ca2+ (e.g., Cavx.y-phenotype), and Cl− channels. Furthermore, we discuss novel genetic defects in K+ channels and associated proteins that underlie distinct epileptic phenotypes in humans, and analyze critically the recent progress in the neurobiology of this disease that has also been provided by investigations on valuable animal models of epilepsy. The abundant and varied lines of evidence discussed here strongly foster assessments for variations in genes encoding for K+ channels and associated proteins in patients with idiopathic epilepsy, provide new avenues for future investigations, and highlight these proteins as critical pharmacological targets.


The Journal of Physiology | 2011

Trigeminal ganglion neuron subtype-specific alterations of CaV2.1 calcium current and excitability in a Cacna1a mouse model of migraine

B Fioretti; Luigi Catacuzzeno; L Sforna; M. B Gerke-Duncan; A.M.J.M. van den Maagdenberg; Fabio Franciolini; Mark Connor; Daniela Pietrobon

Non‐technical summary  Activation of trigeminal neurons innervating the meninges and release of proinflammatory peptides (in particular calcitonin gene‐related peptide (CGRP)) from their terminals are believed to play a key role in generating migraine pain. A monogenic subtype of migraine (familial hemiplegic migraine type‐1 (FHM1)) is caused by gain‐of‐function mutations in a neuronal voltage‐gated calcium channel (Cav2.1) involved in controlling neurotransmitter release from many synaptic terminals including those of trigeminal neurons at the meninges. Using a FHM1 transgenic mouse model, we show that the migraine mutation produces gain‐of‐function (i.e. it increases calcium influx) in a subpopulation of trigeminal neurons that do not innervate the meninges. In contrast, the calcium channels of trigeminal neurons innervating the meninges and releasing CGRP are not affected by the mutation. Congruently, the migraine mutation does not alter CGRP release at the meninges. Our findings suggest that the facilitation of CGRP actions at the meninges does not contribute to the generation of headache in FHM1.


The Journal of Physiology | 2008

The differential expression of low-threshold K+ currents generates distinct firing patterns in different subtypes of adult mouse trigeminal ganglion neurones

Luigi Catacuzzeno; Bernard Fioretti; Daniela Pietrobon; Fabio Franciolini

In adult mouse trigeminal ganglion (TG) neurones we identified three neuronal subpopulations, defined in terms of their firing response to protracted depolarizations, namely MF neurones, characterized by a multiple tonic firing; DMF neurones, characterized by a delay before the beginning of repetitive firing; and SS neurones, characterized by a strongly adapting response. The three subpopulations also differed in several other properties important for defining their functional role in vivo, namely soma size, action potential (AP) shape and capsaicin sensitivity. MF neurones had small soma, markedly long AP and mostly responded to capsaicin, properties typical of a subgroup of C‐type nociceptors. SS neurones had large soma, short AP duration and were mostly capsaicin insensitive, suggesting that most of them have functions other than nociception. DMF neurones were all capsaicin insensitive, had a small soma size and intermediate AP duration, making them functionally distinct from both MF and SS neurones. We investigated the ionic basis underlying the delay to the generation of the first AP of DMF neurones, and the strong adaptation of SS neurones. We found that the expression of a fast‐inactivating, 4‐AP‐ and CP‐339,818‐sensitive K+ current (IA) in DMF neurones plays a critical role in the generation of the delay, whereas a DTX‐sensitive K+ current (IDTX) selectively expressed in SS neurones appeared to be determinant for their strong firing adaptation. A minimal theoretical model of TG neuronal excitability confirmed that IA and IDTX have properties congruent with their suggested role.


Cellular Physiology and Biochemistry | 2006

Expression and Modulation of the Intermediate- Conductance Ca2+-Activated K+ Channel in Glioblastoma GL-15 Cells

Bernard Fioretti; Emilia Castigli; Maria Rita Micheli; Rodolfo Bova; Miriam Sciaccaluga; Alexander A. Harper; Fabio Franciolini; Luigi Catacuzzeno

We report here the expression and properties of the intermediate-conductance Ca2+-activated K+ (IKCa) channel in the GL-15 human glioblastoma cell line. Macroscopic IKCa currents on GL-15 cells displayed a mean amplitude of 7.2±0.8 pA/pF at 0 mV, at day 1 after plating. The current was inhibited by clotrimazole (CTL, IC50=257 nM), TRAM-34 (IC50=55 nM), and charybdotoxin (CTX, IC50=10.3 nM). RT-PCR analysis demonstrated the expression of mRNA encoding the IKCa channel in GL-15 cells. Unitary currents recorded using the inside-out configuration had a conductance of 25 pS, a KD for Ca2+ of 188 nM at -100 mV, and no voltage dependence. We tested whether the IKCa channel expression in GL-15 cells could be the result of an increased ERK activity. Inhibition of the ERK pathway with the MEK antagonist PD98059 (25 µM, for 5 days) virtually suppressed the IKCa current in GL-15 cells. PD98059 treatment also increased the length of cellular processes and up-regulated the astrocytic differentiative marker GFAP. A significant reduction of the IKCa current amplitude was also observed with time in culture, with mean currents of 7.17±0.75 pA/pF at 1-2 days, and 3.11±1.35 pA/pF at 5-6 days after plating. This time-dependent downregulation of the IKCa current was not accompanied by changes in the ERK activity, as assessed by immunoblot analysis. Semiquantitative RT-PCR analysis demonstrated a ~35% reduction of the IKCa channel mRNA resulting from ERK inhibition and a ~50% reduction with time in culture.


Human Molecular Genetics | 2014

Genetically induced dysfunctions of Kir2.1 channels: implications for short QT3 syndrome and autism–epilepsy phenotype

Elena Ambrosini; Federico Sicca; Maria Stefania Brignone; Maria Cristina D'Adamo; Carlo Napolitano; Ilenio Servettini; Francesca Moro; Yanfei Ruan; Luca Guglielmi; Stefania Pieroni; Giuseppe Servillo; Angela Lanciotti; Giulia Valvo; Luigi Catacuzzeno; Fabio Franciolini; Paola Molinari; Maria Marchese; Alessandro Grottesi; Renzo Guerrini; Filippo M. Santorelli; Silvia G. Priori; Mauro Pessia

Short QT3 syndrome (SQT3S) is a cardiac disorder characterized by a high risk of mortality and associated with mutations in Kir2.1 (KCNJ2) channels. The molecular mechanisms leading to channel dysfunction, cardiac rhythm disturbances and neurodevelopmental disorders, potentially associated with SQT3S, remain incompletely understood. Here, we report on monozygotic twins displaying a short QT interval on electrocardiogram recordings and autism–epilepsy phenotype. Genetic screening identified a novel KCNJ2 variant in Kir2.1 that (i) enhanced the channels surface expression and stability at the plasma membrane, (ii) reduced protein ubiquitylation and degradation, (iii) altered protein compartmentalization in lipid rafts by targeting more channels to cholesterol-poor domains and (iv) reduced interactions with caveolin 2. Importantly, our study reveals novel physiological mechanisms concerning wild-type Kir2.1 channel processing by the cell, such as binding to both caveolin 1 and 2, protein degradation through the ubiquitin–proteasome pathway; in addition, it uncovers a potential multifunctional site that controls Kir2.1 surface expression, protein half-life and partitioning to lipid rafts. The reported mechanisms emerge as crucial also for proper astrocyte function, suggesting the need for a neuropsychiatric evaluation in patients with SQT3S and offering new opportunities for disease management.


Journal of Cellular Physiology | 2011

Serum-activated K and Cl currents underlay U87-MG glioblastoma cell migration

Luigi Catacuzzeno; Francesco Aiello; Bernard Fioretti; Luigi Sforna; Emilia Castigli; Paola Ruggieri; Ada Maria Tata; Antonella Calogero; Fabio Franciolini

Glioblastoma cells in vivo are exposed to a variety of promigratory signals, including undefined serum components that infiltrate into high grade gliomas as result of blood–brain barrier breakdown. Glioblastoma cell migration has been further shown to depend heavily on ion channels activity. We have then investigated the modulatory effects of fetal calf serum (FCS) on ion channels, and their involvement in U87‐MG cells migration. Using the perforated patch‐clamp technique we have found that, in a subpopulation of cells (42%), FCS induced: (1) an oscillatory activity of TRAM‐34 sensitive, intermediate‐conductance calcium‐activated K (IKCa) channels, mediated by calcium oscillations previously shown to be induced by FCS in this cell line; (2) a stable activation of a DIDS‐ and NPPB‐sensitive Cl current displaying an outward rectifying instantaneous current‐voltage relationship and a slow, voltage‐dependent inactivation. By contrast, in another subpopulation of cells (32%) FCS induced a single, transient IKCa current activation, always accompanied by a stable activation of the Cl current. The remaining cells did not respond to FCS. In order to understand whether the FCS‐induced ion channel activities are instrumental to promoting cell migration, we tested the effects of TRAM‐34 and DIDS on the FCS‐induced U87‐MG cell migration using transwell migration assays. We found that these inhibitors were able to markedly reduce U87‐MG cell migration in the presence of FCS, and that their co‐application resulted in an almost complete arrest of migration. It is concluded that the modulation of K and Cl ion fluxes is essential for the FCS‐induced glioblastoma cell migration. J. Cell. Physiol. 226: 1926–1933, 2011.


PLOS ONE | 2012

The Inhibition of KCa3.1 Channels Activity Reduces Cell Motility in Glioblastoma Derived Cancer Stem Cells

Paola Ruggieri; Giorgio Mangino; Bernard Fioretti; Luigi Catacuzzeno; Rosa Puca; Donatella Ponti; Massimo Miscusi; Fabio Franciolini; Giuseppe Ragona; Antonella Calogero

In the present study we evaluated the expression of the intermediate conductance calcium-activated potassium (KCa3.1) channel in human glioblastoma stem-like cells (CSCs) and investigated its role in cell motility. While the KCa3.1 channel is not expressed in neuronal- and glial-derived tissues of healthy individuals, both the KCa3.1 mRNA and protein are present in the glioblastoma tumor population, and are significantly enhanced in CSCs derived from both established cell line U87MG and a primary cell line, FCN9. Consistent with these data, voltage-independent and TRAM-34 sensitive potassium currents imputable to the KCa3.1 channel were recorded in the murine GL261 cell line and several primary human glioblastoma cells lines. Moreover, a significantly higher KCa3.1 current was recorded in U87MG-CD133 positive cells as compared to the U87MG-CD133 negative subpopulation. Further, we found that the tumor cell motility is strongly associated with KCa3.1 channel expression. Blockade of the KCa3.1 channel with the specific inhibitor TRAM-34 has in fact a greater impact on the motility of CSCs (reduction of 75%), which express a high level of KCa3.1 channel, than on the FCN9 parental population (reduction of 32%), where the KCa3.1 channel is expressed at lower level. Similar results were also observed with the CSCs derived from U87MG. Because invasion of surrounding tissues is one of the main causes of treatment failure in glioblastoma, these findings can be relevant for future development of novel cancer therapeutic drugs.


American Journal of Physiology-cell Physiology | 2009

Histamine hyperpolarizes human glioblastoma cells by activating the intermediate-conductance Ca2+-activated K+ channel

Bernard Fioretti; Luigi Catacuzzeno; Luigi Sforna; Francesco Aiello; Francesca Pagani; Davide Ragozzino; Emilia Castigli; Fabio Franciolini

The effects of histamine on the membrane potential and currents of human glioblastoma (GL-15) cells were investigated. In perforated whole cell configuration, short (3 s) applications of histamine (100 microM) hyperpolarized the membrane by activating a K(+)-selective current. The response involved the activation of the pyrilamine-sensitive H(1) receptor and Ca(2+) release from thapsigargin-sensitive intracellular stores. The histamine-activated current was insensitive to tetraethylammonium (3 mM), iberiotoxin (100 nM), and d-tubocurarine (100 microM) but was markedly inhibited by charybdotoxin (100 nM), clotrimazole (1 microM), and 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34, 1 microM), a pharmacological profile congruent with the intermediate conductance Ca(2+)-activated K(+) (IK(Ca)) channel. Cell-attached recordings confirmed that histamine activated a K(+) channel with properties congruent with the IK(Ca) channel (voltage independence, 22 pS unitary conductance and slight inward rectification in symmetrical 140 mM K(+)). More prolonged histamine applications (2-3 min) often evoked a sustained IK(Ca) channel activity, which depended on a La(2+) (10 microM)-sensitive Ca(2+) influx. Intracellular Ca(2+) measurements revealed that the sustained IK(Ca) channel activity enhanced the histamine-induced Ca(2+) signal, most likely by a hyperpolarization-induced increase in the driving force for Ca(2+) influx. In virtually all cells examined we also observed the expression of the large conductance Ca(2+)-activated K(+) (BK(Ca)) channel, with a unitary conductance of ca. 230 pS in symmetrical 140 mM K(+), and a Ca(2+) dissociation constant [K(D(Ca))] of ca. 3 microM, at -40 mV. Notably in no instance was the BK(Ca) channel activated by histamine under physiological conditions. The most parsimonious explanation based on the different K(D(Ca)) for the two K(Ca) channels is provided.

Collaboration


Dive into the Fabio Franciolini'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

Miriam Sciaccaluga

Sapienza University of Rome

View shared research outputs
Researchain Logo
Decentralizing Knowledge