Network


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

Hotspot


Dive into the research topics where Franco Tanzi is active.

Publication


Featured researches published by Franco Tanzi.


Stem Cells | 2011

Vascular Endothelial Growth Factor Stimulates Endothelial Colony Forming Cells Proliferation and Tubulogenesis by Inducing Oscillations in Intracellular Ca2+ Concentration†‡§

Silvia Dragoni; Umberto Laforenza; Elisa Bonetti; Francesco Lodola; Cinzia Bottino; Roberto Berra-Romani; Giacomo Carlo Bongio; Maria Pia Cinelli; Germano Guerra; Paolo Pedrazzoli; Vittorio Rosti; Franco Tanzi; Francesco Moccia

Endothelial progenitor cells (EPCs) home from the bone marrow to the site of tissue regeneration and sustain neovascularization after acute vascular injury and upon the angiogenic switch in solid tumors. Therefore, they represent a suitable tool for cell‐based therapy (CBT) in regenerative medicine and provide a novel promising target in the fight against cancer. Intracellular Ca2+ signals regulate numerous endothelial functions, such as proliferation and tubulogenesis. The growth of endothelial colony forming cells (ECFCs), which are EPCs capable of acquiring a mature endothelial phenotype, is governed by store‐dependent Ca2+ entry (SOCE). This study aimed at investigating the nature and the role of VEGF‐elicited Ca2+ signals in ECFCs. VEGF induced asynchronous Ca2+ oscillations, whose latency, amplitude, and frequency were correlated to the growth factor dose. Removal of external Ca2+ (0Ca2+) and SOCE inhibition with N‐(4‐[3,5‐bis(trifluoromethyl)‐1H‐pyrazol‐1‐yl]phenyl)‐4‐methyl‐1,2,3‐thiadiazole‐5‐carboxamide (BTP‐2) reduced the duration of the oscillatory signal. Blockade of phospholipase C‐γ with U73122, emptying the inositol‐1,4,5‐trisphosphate (InsP3)‐sensitive Ca2+ pools with cyclopiazonic acid (CPA), and inhibition of InsP3 receptors with 2‐APB prevented the Ca2+ response to VEGF. VEGF‐induced ECFC proliferation and tubulogenesis were inhibited by the Ca2+‐chelant, BAPTA, and BTP‐2. NF‐κB activation by VEGF was impaired by BAPTA, BTP‐2, and its selective blocker, thymoquinone. Thymoquinone, in turn, suppressed VEGF‐dependent ECFC proliferation and tubulogenesis. These data indicate that VEGF‐induced Ca2+ oscillations require the interplay between InsP3‐dependent Ca2+ release and SOCE, and promote ECFC growth and tubulogenesis by engaging NF‐κB. This novel signaling pathway might be exploited to enhance the outcome of CBT and chemotherapy. STEM CELLS 2011;29:1898–1907


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.


Current Medicinal Chemistry | 2012

Store-Dependent Ca2+ Entry in Endothelial Progenitor Cells As a Perspective Tool to Enhance Cell-Based Therapy and Adverse Tumour Vascularization

Francesco Moccia; Silvia Dragoni; F. Lodola; E. Bonetti; Cinzia Bottino; Germano Guerra; Umberto Laforenza; Vittorio Rosti; Franco Tanzi

Endothelial progenitor cells (EPCs) have recently been employed in cell-based therapy (CBT) to promote neovascularization and regeneration of ischemic organs, such as heart and limbs. Furthermore, EPCs may be recruited from bone marrow by growing tumors to drive the angiogenic switch through physical engrafting into the lumen of nascent vessels or paracrine release of pro-angiogenic factors. CBT is hampered by the paucity of EPCs harvested from peripheral blood and suffered from several pitfalls, including the differentiation outcome of transplanted cells and low percentage of engrafted cells. Therefore, CBT will benefit from a better understanding of the signal transduction pathway(s) which govern(s) EPC homing, proliferation and incorporation into injured tissues. At the same time, this information might outline alternative molecular targets to combat tumoral neovascularization. We have recently found that store-operated Ca(2+) entry, a Ca(2+)-permeable membrane pathway that is activated upon depletion of the inositol-1,4,5-trisphosphate-sensitive Ca(2+) pool, is recruited by vascular endothelial growth factor to support proliferation and tubulogenesis in human circulating endothelial colony forming cells (ECFCs). ECFCs are a subgroup of EPCs that circulate in the peripheral blood of adult individuals and are able to proliferate and differentiate into endothelial cells and form capillary networks in vitro and contribute to neovessel formation in vivo. The present review will discuss the relevance of SOCE to ECFC-based cell therapy and will address the pharmacological inhibition of store-dependent Ca(2+) channels as a promising target for anti-angiogenic treatments.


Stem Cells and Development | 2010

Store-Operated Ca2+ Entry Is Expressed in Human Endothelial Progenitor Cells

Yuly Sánchez-Hernández; Umberto Laforenza; Elisa Bonetti; Jacopo M. Fontana; Silvia Dragoni; Marika A. Russo; José Everardo Avelino-Cruz; Sergio Schinelli; Domenico Testa; Germano Guerra; Vittorio Rosti; Franco Tanzi; Francesco Moccia

Endothelial progenitor cells (EPCs) may be recruited from the bone marrow to sites of tissue regeneration to sustain neovascularization and reendothelialization after acute vascular injury. This feature makes them particularly suitable for cell-based therapy. In mature endothelium, store-operated Ca(2+) entry (SOCE) is activated following emptying of inositol-1,4,5-trisphosphate-sensitive stores, and controls a wide number of functions, including proliferation, nitric oxide synthesis, and vascular permeability. The present work aimed at investigating SOCE expression in EPCs harvested from both peripheral blood (PB-EPCs) and umbilical cord blood (UCB-EPCs) by employing both Ca(2+) imaging and molecular biology techniques. SOCE was induced upon either pharmacological (ie, cyclopiazonic acid) or physiological (ie, ATP) depletion of the intracellular Ca(2+) pool. Further, store-dependent Ca(2+) entry was inhibited by the SOCE inhibitor, N-(4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl)-4-methyl-1,2,3-thiadiazole-5-carboxamide (BTP-2). Real-time reverse transcription-polymerase chain reaction and western blot analyses showed that both PB-EPCs and UCB-EPCs express all the molecular candidates to mediate SOCE in differentiated cells, including TRPC1, TRPC4, Orai1, and Stim1. Moreover, pharmacological maneuvers demonstrated that, as well as in differentiated endothelial cells, the signal transduction pathway leading to depletion of the intracellular Ca(2+) pool impinged on the phospholipase C/inositol-1,4,5-trisphosphate pathway. Finally, blockage of SOCE with BTP-2 impaired PB-EPC proliferation. These findings provide the first evidence that EPCs express SOCE, which might thus be regarded as a novel target to enhance the regenerative outcome of cell-based therapy.


Frontiers in Cellular Neuroscience | 2015

Stim and Orai proteins in neuronal Ca2+ signaling and excitability

Francesco Moccia; Estella Zuccolo; Teresa Soda; Franco Tanzi; Germano Guerra; Lisa Mapelli; Francesco Lodola; Egidio D’Angelo

Stim1 and Orai1 are ubiquitous proteins that have long been known to mediate Ca2+ release-activated Ca2+ (CRAC) current (ICRAC) and store-operated Ca2+ entry (SOCE) only in non-excitable cells. SOCE is activated following the depletion of the endogenous Ca2+ stores, which are mainly located within the endoplasmic reticulum (ER), to replete the intracellular Ca2+ reservoir and engage specific Ca2+-dependent processes, such as proliferation, migration, cytoskeletal remodeling, and gene expression. Their paralogs, Stim2, Orai2 and Orai3, support SOCE in heterologous expression systems, but their physiological role is still obscure. Ca2+ inflow in neurons has long been exclusively ascribed to voltage-operated and receptor-operated channels. Nevertheless, recent work has unveiled that Stim1–2 and Orai1-2, but not Orai3, proteins are also expressed and mediate SOCE in neurons. Herein, we survey current knowledge about the neuronal distribution of Stim and Orai proteins in rodent and human brains; we further discuss that Orai2 is the main pore-forming subunit of CRAC channels in central neurons, in which it may be activated by either Stim1 or Stim2 depending on species, brain region and physiological stimuli. We examine the functions regulated by SOCE in neurons, where this pathway is activated under resting conditions to refill the ER, control spinogenesis and regulate gene transcription. Besides, we highlighted the possibility that SOCE also controls neuronal excitation and regulate synaptic plasticity. Finally, we evaluate the involvement of Stim and Orai proteins in severe neurodegenerative and neurological disorders, such as Alzheimer’s disease and epilepsy.


World Journal of Biological Chemistry | 2012

Update on vascular endothelial Ca(2+) signalling: A tale of ion channels, pumps and transporters.

Francesco Moccia; Roberto Berra-Romani; Franco Tanzi

A monolayer of endothelial cells (ECs) lines the lumen of blood vessels and forms a multifunctional transducing organ that mediates a plethora of cardiovascular processes. The activation of ECs from as state of quiescence is, therefore, regarded among the early events leading to the onset and progression of potentially lethal diseases, such as hypertension, myocardial infarction, brain stroke, and tumor. Intracellular Ca(2+) signals have long been know to play a central role in the complex network of signaling pathways regulating the endothelial functions. Notably, recent work has outlined how any change in the pattern of expression of endothelial channels, transporters and pumps involved in the modulation of intracellular Ca(2+) levels may dramatically affect whole body homeostasis. Vascular ECs may react to both mechanical and chemical stimuli by generating a variety of intracellular Ca(2+) signals, ranging from brief, localized Ca(2+) pulses to prolonged Ca(2+) oscillations engulfing the whole cytoplasm. The well-defined spatiotemporal profile of the subcellular Ca(2+) signals elicited in ECs by specific extracellular inputs depends on the interaction between Ca(2+) releasing channels, which are located both on the plasma membrane and in a number of intracellular organelles, and Ca(2+) removing systems. The present article aims to summarize both the past and recent literature in the field to provide a clear-cut picture of our current knowledge on the molecular nature and the role played by the components of the Ca(2+) machinery in vascular ECs under both physiological and pathological conditions.


Current Vascular Pharmacology | 2014

Ca 2+ Signalling in Endothelial Progenitor Cells: A Novel Means to Improve Cell-Based Therapy and Impair Tumour Vascularisation

Francesco Moccia; Francesco Lodola; Silvia Dragoni; Elisa Bonetti; Cinzia Bottino; Germano Guerra; Umberto Laforenza; Vittorio Rosti; Franco Tanzi

Endothelial progenitor cells (EPCs) have recently been employed in cell-based therapy (CBT) to promote regeneration of ischemic organs, such as heart and limbs. Furthermore, EPCs may sustain tumour vascularisation and provide an additional target for anticancer therapies. CBT is limited by the paucity of cells harvested from peripheral blood and suffers from several pitfalls, including the low rate of engrafted EPCs, whereas classic antiangiogenic treatments manifest a number of side effects and may induce resistance into the patients. CBT will benefit of a better understanding of the signal transduction pathway(s) which drive(s) EPC proliferation, trafficking, and incorporation into injured tissues. At the same time, this information might outline alternative molecular targets to impair tumor neovascularisation and improve the therapeutic outcome of antiangiogenic strategies. An increase in intracellular Ca(2+) concentration is the key signal in the regulation of cellular replication, migration, and differentiation. In particular, Ca(2+) signalling may regulate cellcycle progression, due to the Ca(2+)-sensitivity of a number of cycline-dependent kinases, and gene expression, owing to the Ca(2+)-dependence of several transcription factors. Recent work has outlined the role of the so-called store-operated Ca(2+) entry in driving EPC proliferation and migration. Unravelling the mechanisms guiding EPC engraftment into neovessels might supply the biological bases required to improve CBT and anticancer treatments. For example, genetic manipulation of the Ca(2+) signalling machinery could provide a novel approach to increase the extent of limb regeneration or preventing tumour vascularisation by EPCs.


Stem Cells and Development | 2013

Canonical Transient Receptor Potential 3 Channel Triggers Vascular Endothelial Growth Factor-Induced Intracellular Ca2+ Oscillations in Endothelial Progenitor Cells Isolated from Umbilical Cord Blood

Silvia Dragoni; Umberto Laforenza; Elisa Bonetti; Francesco Lodola; Cinzia Bottino; Germano Guerra; Alessandro Borghesi; Mauro Stronati; Vittorio Rosti; Franco Tanzi; Francesco Moccia

Endothelial colony-forming cells (ECFCs) are the only endothelial progenitor cells (EPCs) that are capable of acquiring a mature endothelial phenotype. ECFCs are mainly mobilized from bone marrow to promote vascularization and represent a promising tool for cell-based therapy of severe ischemic diseases. Vascular endothelial growth factor (VEGF) stimulates the proliferation of peripheral blood-derived ECFCs (PB-ECFCs) through oscillations in intracellular Ca(2+) concentration ([Ca(2+)]i). VEGF-induced Ca(2+) spikes are driven by the interplay between inositol-1,4,5-trisphosphate (InsP3)-dependent Ca(2+) release and store-operated Ca(2+) entry (SOCE). The therapeutic potential of umbilical cord blood-derived ECFCs (UCB-ECFCs) has also been shown in recent studies. However, VEGF-induced proliferation of UCB-ECFCs is faster compared with their peripheral counterpart. Unlike PB-ECFCs, UCB-ECFCs express canonical transient receptor potential channel 3 (TRPC3) that mediates diacylglycerol-dependent Ca(2+) entry. The present study aimed at investigating whether the higher proliferative potential of UCB-ECFCs was associated to any difference in the molecular underpinnings of their Ca(2+) response to VEGF. We found that VEGF induces oscillations in [Ca(2+)]i that are patterned by the interaction between InsP3-dependent Ca(2+) release and SOCE. Unlike PB-ECFCs, VEGF-evoked Ca(2+) oscillations do not arise in the absence of extracellular Ca(2+) entry and after pharmacological (with Pyr3 and flufenamic acid) and genetic (by employing selective small interference RNA) suppression of TRPC3. VEGF-induced UCB-ECFC proliferation is abrogated on inhibition of the intracellular Ca(2+) spikes. Therefore, the Ca(2+) response to VEGF in UCB-ECFCs is shaped by a different Ca(2+) machinery as compared with PB-ECFCs, and TRPC3 stands out as a promising target in EPC-based treatment of ischemic pathologies.


Neuroscience | 2003

Dual effect of Zn2+ on multiple types of voltage-dependent Ca2+ currents in rat palaeocortical neurons.

Jacopo Magistretti; Loretta Castelli; Vanni Taglietti; Franco Tanzi

The effects of Zn(2+) were evaluated on high-voltage-activated Ca(2+) currents expressed by pyramidal neurons acutely dissociated from rat piriform cortex. Whole-cell, patch-clamp experiments were carried out using Ba(2+) (5 mM) as the charge carrier. Zn(2+) blocked total high-voltage-activated Ba(2+) currents with an IC(50) of approximately 21 microM. In addition, after application of non-saturating Zn(2+) concentrations, residual currents activated with substantially slower kinetics than control Ba(2+) currents. Both of the above-mentioned effects of Zn(2+) were also observed in high-voltage-activated currents recorded in the presence of nearly-physiological concentrations of extracellular Ca(2+) (1 and 2 mM) rather than Ba(2+). Under the latter conditions, 30 microM Zn(2+) inhibited high-voltage-activated currents somewhat less than observed in extracellular Ba(2+) (approximately 47% and approximately 41%, respectively, vs. approximately 59%), but slowed Ca(2+)-current activation to very similar degrees. All of the pharmacological components in which Ba(2+) currents could be dissected (L-, N-, P/Q-, and R-type) were inhibited by Zn(2+), the percentage of current blocked by 30 microM Zn(2+) ranging from 34 to 57%. Moreover, the activation kinetics of all pharmacological Ba(2+) current components were slowed by Zn(2+). Hence, the lower activation speed observed in residual Ba(2+) currents after Zn(2+) block is due to a true slowing of macroscopic Ca(2+)-current activation kinetics and not to the preferential inhibition of a fast-activating current component. The inhibitory effect of Zn(2+) on Ba(2+) current amplitude was voltage-independent over the whole voltage range explored (-60 to +30 mV), hence the Zn(2+)-dependent decrease of Ba(2+) current activation speed is not the consequence of a voltage- and time-dependent relief from block. Zn(2+) also caused a slight, but significant, reduction of Ba(2+) current deactivation speed upon repolarization, which is further evidence against a depolarization-dependent unblocking mechanism. Finally, the slowing effect of Zn(2+) on Ca(2+)-channel activation kinetics was found to result in a significant, extra reduction of Ba(2+) current amplitude when action-potential-like waveforms, rather than step pulses, were used as depolarizing stimuli. We conclude that Zn(2+) exerts a dual action on multiple types of voltage-gated Ca(2+) channels, causing a blocking effect and altering the speed at which channels are delivered to conducting states, with mechanism(s) that could be distinct.


Journal of Cellular Physiology | 2003

Epidermal growth factor induces intracellular Ca2+ oscillations in microvascular endothelial cells

Francesco Moccia; Roberto Berra-Romani; Simona Tritto; Silvia Signorelli; Vanni Taglietti; Franco Tanzi

An increase in intracellular Ca2+ concentration ([Ca2+]i) may play a role in the proliferative effect of several growth factors. In this study, the changes in [Ca2+]i elicited by epidermal growth factor (EGF) in rat cardiac microvascular endothelial cells (CMEC) have been investigated by using fura‐2 conventional and confocal microscopy. A large heterogeneity in the latency and in the pattern of the Ca2+ response was found at each dose of EGF (2.5–100 ng/ml), whereas some cells displayed a non‐oscillatory behavior and others exhibited a variable number of Ca2+ oscillations. On average, the fraction of responsive cells, the total number of oscillations and the duration of the Ca2+ signal were higher at around 10 ng/ml EGF, while there was no dose‐dependence in the lag time and in the amplitude of the [Ca2+]i increase. EGF‐induced Ca2+ spikes were abolished by the tyrosine kinase inhibitor genistein, but not by its inactive analogue daidzein, and by the phospholipase C blocker NCDC. Only 1–2 transients could be elicited in Ca2+‐free solution, while re‐addition of extracellular Ca2+ recovered the spiking activity. The oscillatory signal was prevented by the SERCA inhibitor thapsigargin and abolished by the calcium entry blockers Ni2+ and La3+. Moreover, EGF‐induced Ca2+ transients were abolished by the InsP3 receptor blocker caffeine, while ryanodine was without effect. Confocal imaging microscopy showed that the Ca2+ response to EGF was localized both in the cytoplasm and in the nucleus. We suggest that EGF‐induced [Ca2+]i increase may play a role in the proliferative action of EGF on endothelial cells.

Collaboration


Dive into the Franco Tanzi'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