Network


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

Hotspot


Dive into the research topics where Armando A. Genazzani is active.

Publication


Featured researches published by Armando A. Genazzani.


Cell | 2002

NAADP Mobilizes Ca2+ from Reserve Granules, Lysosome-Related Organelles, in Sea Urchin Eggs

Grant C. Churchill; Yuhei Okada; Justyn M. Thomas; Armando A. Genazzani; Sandip Patel; Antony Galione

Abstract Nicotinic acid adenine dinucleotide phosphate (NAADP) mobilizes Ca 2+ in many cells and species. Unlike other Ca 2+ -mobilizing messengers, NAADP mobilizes Ca 2+ from an unknown store that is not the endoplasmic reticulum, the store traditionally associated with messenger-mediated Ca 2+ signaling. Here, we demonstrate the presence of a Ca 2+ store in sea urchin eggs mobilized by NAADP that is dependent on a proton gradient maintained by an ATP-dependent vacuolar-type proton pump. Moreover, we provide pharmacological and biochemical evidence that this Ca 2+ store is the reserve granule, the functional equivalent of a lysosome in the sea urchin egg. These findings represent an unsuspected mechanism for messenger-mediated Ca 2+ release from lysosome-related organelles.


Current Biology | 2003

Sperm Deliver a New Second Messenger: NAADP

Grant C. Churchill; John S. O'Neill; Roser Masgrau; Sandip Patel; Justyn M. Thomas; Armando A. Genazzani; Antony Galione

NAADP is a highly potent mobilizer of Ca(2+), which in turn triggers Ca(2+)-induced Ca(2+) release pathways in a wide range of species. Nevertheless, NAADP is not presently classified as a second messenger because it has not been shown to increase in response to a physiological stimulus. We now report a dramatic increase in NAADP during sea urchin egg fertilization that was largely due to production in sperm upon contacting egg jelly. The NAADP bolus plays a physiological role upon delivery to the egg based on its ability to induce a cortical flash, a depolarization-induced activation of L-type Ca(2+) channels. Moreover, the sperm-induced cortical flash was eliminated in eggs desensitized to NAADP. We conclude that an NAADP increase plays a physiologically relevant role during fertilization and provides the first conclusive demonstration that NAADP is a genuine second messenger.


Current Biology | 1999

Nicotinic acid adenine dinucleotide phosphate triggers Ca2+ release from brain microsomes

Judit Bak; Peter R. White; György Timár; Ludwig Missiaen; Armando A. Genazzani; Antony Galione

Mobilization of Ca2+ from intracellular stores is an important mechanism for generating cytoplasmic Ca2+ signals [1]. Two families of intracellular Ca(2+)-release channels - the inositol-1,4, 5-trisphosphate (IP3) receptors and the ryanodine receptors (RyRs) - have been described in mammalian tissues [2]. Recently, nicotinic acid adenine dinucleotide phosphate (NAADP), a molecule derived from NADP+, has been shown to trigger Ca2+ release from intracellular stores in invertebrate eggs [3] [4] [5] [6] and pancreatic acinar cells [7]. The nature of NAADP-induced Ca2+ release is unknown but it is clearly distinct from the IP3- and cyclic ADP ribose (cADPR)-sensitive mechanisms in eggs (reviewed in [8] [9]). Furthermore, mammalian cells can synthesize and degrade NAADP, suggesting that NAADP-induced Ca2+ release may be widespread and thus contribute to the complexity of Ca2+ signalling [10] [11]. Here, we show for the first time that NAADP evokes Ca2+ release from rat brain microsomes by a mechanism that is distinct from those sensitive to IP3 or cADPR, and has a remarkably similar pharmacology to the action of NAADP in sea urchin eggs [12]. Membranes prepared from the same rat brain tissues are able to support the synthesis and degradation of NAADP. We therefore suggest that NAADP-mediated Ca2+ signalling could play an important role in neuronal Ca2+ signalling.


Journal of Cell Biology | 2009

WldS protein requires Nmnat activity and a short N-terminal sequence to protect axons in mice

Laura Conforti; Anna L. Wilbrey; Giacomo Morreale; Lucie Janeckova; Bogdan Beirowski; Robert Adalbert; Francesca Mazzola; Michele Di Stefano; Robert Hartley; Elisabetta Babetto; Trevor Stanley Smith; Jonathan Gilley; Richard A. Billington; Armando A. Genazzani; Richard R. Ribchester; Giulio Magni; Michael P. Coleman

The slow Wallerian degeneration (WldS) protein protects injured axons from degeneration. This unusual chimeric protein fuses a 70–amino acid N-terminal sequence from the Ube4b multiubiquitination factor with the nicotinamide adenine dinucleotide–synthesizing enzyme nicotinamide mononucleotide adenylyl transferase 1. The requirement for these components and the mechanism of WldS-mediated neuroprotection remain highly controversial. The Ube4b domain is necessary for the protective phenotype in mice, but precisely which sequence is essential and why are unclear. Binding to the AAA adenosine triphosphatase valosin-containing protein (VCP)/p97 is the only known biochemical property of the Ube4b domain. Using an in vivo approach, we show that removing the VCP-binding sequence abolishes axon protection. Replacing the WldS VCP-binding domain with an alternative ataxin-3–derived VCP-binding sequence restores its protective function. Enzyme-dead WldS is unable to delay Wallerian degeneration in mice. Thus, neither domain is effective without the function of the other. WldS requires both of its components to protect axons from degeneration.


British Journal of Pharmacology | 2008

Inhibitors of histone deacetylase (HDAC) restore the p53 pathway in neuroblastoma cells

Fabrizio Condorelli; I Gnemmi; A Vallario; Armando A. Genazzani; Pier Luigi Canonico

Inhibitors of histone deacetylase (HDAC) are emerging as a promising class of anti‐cancer drugs, but a generic deregulation of transcription in neoplastic cells cannot fully explain their therapeutic effects. In this study we evaluated alternative molecular mechanisms by which HDAC inhibitors could affect neuroblastoma viability.


PLOS ONE | 2012

Store-operated Ca2+ entry is remodelled and controls in vitro angiogenesis in endothelial progenitor cells isolated from tumoral patients.

Francesco Lodola; Umberto Laforenza; Elisa Bonetti; Dmitry Lim; Silvia Dragoni; Cinzia Bottino; Hwei Ling Ong; Germano Guerra; Carlo Ganini; Margherita Massa; Mariangela Manzoni; Indu S. Ambudkar; Armando A. Genazzani; Vittorio Rosti; Paolo Pedrazzoli; Franco Tanzi; Francesco Moccia; Camillo Porta

Background Endothelial progenitor cells (EPCs) may be recruited from bone marrow to sustain tumor vascularisation and promote the metastatic switch. Understanding the molecular mechanisms driving EPC proliferation and tubulogenesis could outline novel targets for alternative anti-angiogenic treatments. Store-operated Ca2+ entry (SOCE), which is activated by a depletion of the intracellular Ca2+ pool, regulates the growth of human EPCs, where is mediated by the interaction between the endoplasmic reticulum Ca2+-sensor, Stim1, and the plasmalemmal Ca2+ channel, Orai1. As oncogenesis may be associated to the capability of tumor cells to grow independently on Ca2+ influx, it is important to assess whether SOCE regulates EPC-dependent angiogenesis also in tumor patients. Methodology/Principal Findings The present study employed Ca2+ imaging, recombinant sub-membranal and mitochondrial aequorin, real-time polymerase chain reaction, gene silencing techniques and western blot analysis to investigate the expression and the role of SOCE in EPCs isolated from peripheral blood of patients affected by renal cellular carcinoma (RCC; RCC-EPCs) as compared to control EPCs (N-EPCs). SOCE, activated by either pharmacological (i.e. cyclopiazonic acid) or physiological (i.e. ATP) stimulation, was significantly higher in RCC-EPCs and was selectively sensitive to BTP-2, and to the trivalent cations, La3+ and Gd3+. Furthermore, 2-APB enhanced thapsigargin-evoked SOCE at low concentrations, whereas higher doses caused SOCE inhibition. Conversely, the anti-angiogenic drug, carboxyamidotriazole (CAI), blocked both SOCE and the intracellular Ca2+ release. SOCE was associated to the over-expression of Orai1, Stim1, and transient receptor potential channel 1 (TRPC1) at both mRNA and protein level The intracellular Ca2+ buffer, BAPTA, BTP-2, and CAI inhibited RCC-EPC proliferation and tubulogenesis. The genetic suppression of Stim1, Orai1, and TRPC1 blocked CPA-evoked SOCE in RCC-EPCs. Conclusions SOCE is remodelled in EPCs from RCC patients and stands out as a novel molecular target to interfere with RCC vascularisation due to its ability to control proliferation and tubulogenesis.


Autophagy | 2008

NAD depletion by FK866 induces autophagy.

Richard A. Billington; Armando A. Genazzani; Cristina Travelli; Fabrizio Condorelli

NAD is a multifunctional molecule involved in both metabolic processes and signalling pathways. Such signalling pathways consume NAD which is replenished via one of several biosynthesis pathways. We show that influx of NAD across the plasma membrane may be able to contribute to the homeostasis of intracellular NAD levels. Indeed, extracellular application of NAD was able to replete NAD levels that had been lowered pharmacologically using the novel drug FK866 and was also able to rescue cells from FK866-induced cell death. A marked lag between the drop in NAD levels and cell death prompted us to investigate the mechanism of cell death. We were unable to find evidence of apoptosis as assessed by immunoblotting for the Caspase 3 activation fragment and immunostaining for cytochrome C and AIF translocation. We, therefore, investigated whether autophagy was initiated by FK866. Indeed, we were able to observe the formation of LC3-positive vesicles that had fused with lysosomes in FK866-treated but not control cells. Furthermore, this autophagic phenotype could be reverted by the addition of NAD to the extracellular medium. Addendum to: Billington RA, Travelli C, Ercolano E, Galli U, Blasi Roman C, Grolla AA, Canonico PL, Condorelli F, Genazzani AA. Characterization of NAD uptake in mammalian cells. J Biol Chem 2008; In Press.


Journal of Biological Chemistry | 2008

Characterization of NAD Uptake in Mammalian Cells

Richard A. Billington; Cristina Travelli; Emanuela Ercolano; Ubaldina Galli; Cintia R. Blasi Roman; Ambra A. Grolla; Pier Luigi Canonico; Fabrizio Condorelli; Armando A. Genazzani

Recent evidence has shown that NAD(P) plays a variety of roles in cell-signaling processes. Surprisingly, the presence of NAD(P) utilizing ectoenzymes suggests that NAD(P) is present extracellularly. Indeed, nanomolar concentrations of NAD have been found in plasma and other body fluids. Although very high concentrations of NAD have been shown to enter cells, it is not known whether lower, more physiological concentrations are able to be taken up. Here we show that two mammalian cell types are able to transport low NAD concentrations effectively. Furthermore, extracellular application of NAD was able to counteract FK866-induced cell death and restore intracellular NAD(P) levels. We propose that NAD uptake may play a role in physiological NAD homeostasis.


Current Biology | 2001

NAADP receptors are present and functional in the heart

Judit Bak; Richard A. Billington; Gyoergy Timar; Alice C. Dutton; Armando A. Genazzani

Alongside the well-studied inositol 1,4,5 trisphosphate and ryanodine receptors, evidence is gathering that a new intracellular release mechanism, gated by the pyridine nucleotide nicotinic acid adenine dinucleotide phosphate (NAADP), is present in numerous organisms, ranging from plant to mammalian cells (reviewed in [1]). Most cells have been shown to express at least two Ca(2+)-release mechanisms controlled by different messengers, and this can lead to redundancy, convergence, or divergence of responses. One exception appears to be muscle and heart contractile tissues. Here, it is thought that the dominant intracellular channel is the ryanodine receptor, while IP(3) receptors are poorly expressed and their role appears to be negligible. We now report that NAADP receptors are functional and abundant in cardiac microsomes. NAADP binds specifically and with high affinity (130 pM and 4 nM) to two sites on cardiac microsomes and releases Ca(2+) with an apparent EC(50) of 323 +/- 14 nM. Furthermore, binding experiments show that this receptor displays both positive and negative cooperativity, a peculiarity unique among intracellular Ca(2+) channels. Therefore, we show that the heart possesses multiple mechanisms to increase the complexity of Ca(2+) signaling and that NAADP may be integral in the functioning of this organ.


The FASEB Journal | 2002

A pivotal role for cADPR-mediated Ca2+ signaling: regulation of endothelin-induced contraction in peritubular smooth muscle cells

Fortunata Barone; Armando A. Genazzani; Antonio Conti; Grant C. Churchill; Fioretta Palombi; Elio Ziparo; Vincenzo Sorrentino; Antony Galione; Antonio Filippini

cADPR, a potent calcium‐mobilizing intracellular messenger synthesized by ADP‐ribosyl cyclases regulates openings of ryanodine receptors (RyR). Here we report that in the rat testis, a functional cADPR Ca2+ release system is essential for the contractile response of peritubular smooth muscle cells (PSMC) to endothelin (ET). We previously showed that this potent smooth muscle agonist elicits intracellular Ca2+ release in PSMC and seminiferous tubule contraction via activation of ETA and ETB receptors. ETB‐R induces the mobilization of a thapsigargin‐sensitive but IP3‐independent intracellular Ca2+ pool. Stimulation of permeabilized PSMC with cADPR was found to elicit large Ca2+ releases blocked by either a selective antagonist of cADPR or a RyR blocker, but not by heparin. Western blotting and confocal fluorescence microscopy indicated the specific expression of type 2 RyR in perinuclear localization. ET was found to stimulate the activity of ADP‐ribosyl cyclase. Microinjection of the selective cADPR antagonist 8NH2‐cADPR completely abolished subsequent stimulation of Ca2+ signaling via ETA and ETB receptors. cADPR therefore appears to have an obligatory role for ETA‐R and ETB‐R‐mediated calcium signaling in PSMC. However, ETB‐R seem to be coupled exclusively to cADPR whereas ETA‐R activation may be linked to IP3 and cADPR signaling pathways.

Collaboration


Dive into the Armando A. Genazzani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gian Cesare Tron

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Dmitry Lim

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guido Cavaletti

University of Milano-Bicocca

View shared research outputs
Top Co-Authors

Avatar

Salvatore Terrazzino

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge