Network


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

Hotspot


Dive into the research topics where Francesca Cencetti is active.

Publication


Featured researches published by Francesca Cencetti.


Molecular Biology of the Cell | 2010

Transforming Growth Factor-β1 Induces Transdifferentiation of Myoblasts into Myofibroblasts via Up-Regulation of Sphingosine Kinase-1/S1P3 Axis

Francesca Cencetti; Caterina Bernacchioni; Paola Nincheri; Chiara Donati; Paola Bruni

Transforming growth factor-β1 induces Smad-dependent transdifferentiation of myoblasts into myofibroblasts via up-regulation of sphingosine kinase-1/S1P3 axis with a mechanism involving Rho/Rho kinase activation.


Stem Cells | 2007

Sphingosine 1‐Phosphate Mediates Proliferation and Survival of Mesoangioblasts

Chiara Donati; Francesca Cencetti; Paola Nincheri; Caterina Bernacchioni; Silvia Brunelli; Emilio Clementi; Giulio Cossu; Paola Bruni

Mesoangioblasts are stem cells capable of differentiating in various mesodermal tissues and are presently regarded as suitable candidates for cell therapy of muscle degenerative diseases, as well as myocardial infarction. The enhancement of their proliferation and survival after injection in vivo could greatly improve their ability to repopulate damaged tissues. In this study, we show that the bioactive sphingolipid sphingosine 1‐phosphate (S1P) regulates critical functions of mesoangioblast cell biology. S1P evoked a full mitogenic response in mesoangioblasts, measured by labeled thymidine incorporation and cell counting. Moreover, S1P strongly counteracted the apoptotic process triggered by stimuli as diverse as serum deprivation, C2‐ceramide treatment, or staurosporine treatment, as assessed by cell counting, as well as histone‐associated fragments and caspase‐3 activity determinations. S1P acts both as an intracellular messenger and through specific membrane receptors. Real‐time polymerase chain reaction analysis revealed that mesoangioblasts express the S1P‐specific receptor S1P3 and, to a minor extent, S1P1 and S1P2. By using S1P receptor subtype‐specific agonists and antagonists, we found that the proliferative response to S1P was mediated mainly by S1P2. By contrast, the antiapoptotic effect did not implicate S1P receptors. These findings demonstrate an important role of S1P in mesoangioblast proliferation and survival and indicate that targeting modulation of S1P‐dependent signaling pathways may be used to improve the efficiency of muscle repair by these cells.


Journal of Cellular Physiology | 2008

Sphingosine kinase activity is required for myogenic differentiation of C2C12 myoblasts

Elisabetta Meacci; Francesca Nuti; Chiara Donati; Francesca Cencetti; Marta Farnararo; Paola Bruni

Sphingosine kinase (SphK) is a conserved lipid kinase that catalyzes the formation of sphingosine 1‐phosphate (S1P), an important lipid mediator, which regulates fundamental biological processes. Here, we provide evidence that SphK is required for the achievement of cell growth arrest as well as myogenic differentiation of C2C12 myoblasts. Indeed, SphK activity, SphK1 protein content and S1P formation were found to be enhanced in myoblasts that became confluent as well as in differentiating cells. Enforced expression of SphK1 reduced the myoblast proliferation rate, enhanced the expression of myogenic differentiation markers and anticipated the onset of differentiated muscle phenotype. Conversely, down‐regulation of SphK1 by specific silencing by RNA interference or overexpression of the catalytically inactive SphK1, significantly increased cell growth and delayed the beginning of myogenesis; noticeably, exogenous addition of S1P rescued the biological processes. Importantly, stimulation of myogenesis in SphK1‐overexpressing myoblasts was abrogated by treatment with short interfering RNA specific for S1P2 receptor. This is the first report of the role of endogenous SphK1 in myoblast growth arrest and stimulation of myogenesis through the formation of S1P that acts as morphogenic factor via the engagement of S1P2. J. Cell. Physiol. 214:210–220, 2008.


Biochimie | 2012

Ceramide 1-phosphate stimulates proliferation of C2C12 myoblasts

Patricia Gangoiti; Caterina Bernacchioni; Chiara Donati; Francesca Cencetti; Alberto Ouro; Antonio Gómez-Muñoz; Paola Bruni

Recent studies have established specific cellular functions for different bioactive sphingolipids in skeletal muscle cells. Ceramide 1-phosphate (C1P) is an important bioactive sphingolipid that has been involved in cell growth and survival. However its possible role in the regulation of muscle cell homeostasis has not been so far investigated. In this study, we show that C1P stimulates myoblast proliferation, as determined by measuring the incorporation of tritiated thymidine into DNA, and progression of the myoblasts through the cell cycle. C1P induced phosphorylation of glycogen synthase kinase-3β and the product of retinoblastoma gene, and enhanced cyclin D1 protein levels. The mitogenic action of C1P also involved activation of phosphatidylinositol 3-kinase/Akt, ERK1/2 and the mammalian target of rapamycin. These effects of C1P were independent of interaction with a putative Gi-coupled C1P receptor as pertussis toxin, which maintains Gi protein in the inactive form, did not affect C1P-stimulated myoblast proliferation. By contrast, C1P was unable to inhibit serum starvation- or staurosporine-induced apoptosis in the myoblasts, and did not affect myogenic differentiation. Collectively, these results add up to the current knowledge on cell types targeted by C1P, which so far has been mainly confined to fibroblasts and macrophages, and extend on the mechanisms by which C1P exerts its mitogenic effects. Moreover, the biological activities of C1P described in this report establish that this phosphosphingolipid may be a relevant cue in the regulation of skeletal muscle regeneration, and that C1P-metabolizing enzymes might be important targets for developing cellular therapies for treatment of skeletal muscle degenerative diseases, or tissue injury.


Biochimica et Biophysica Acta | 2012

Sphingosine 1-phosphate stimulates proliferation and migration of satellite cells Role of S1P receptors.

Serena Calise; Sabrina Blescia; Francesca Cencetti; Caterina Bernacchioni; Chiara Donati; Paola Bruni

Satellite cells are resident stem cells of skeletal muscle; they are normally quiescent but upon post-trauma activation start to proliferate and fuse with damaged fibers contributing to muscle regeneration. In this study the effect of the bioactive sphingolipid sphingosine 1-phosphate (S1P) on the proliferative and migratory response of murine satellite cells has been examined. S1P was found to stimulate labeled thymidine incorporation in a phosphatidylinositol 3-kinase-dependent manner. Moreover, by employing selective S1P receptor agonists and antagonists and silencing individual S1P receptors, the mitogenic action of S1P in satellite cells was shown to depend on S1P2 and S1P3. Notably, by using different experimental approaches S1P was found to positively influence satellite cell migration, necessary for their recruitment at the site of muscle damage. Interestingly, the specific silencing of individual S1P receptor subtypes demonstrated the pivotal role of S1P1 and S1P4 in mediating the S1P migratory effect. This latter result demonstrates for the first time that S1P4 receptor has a role in skeletal muscle cells, supporting the notion that this receptor subtype plays a biological action broader than that so far identified in lymphoid tissue. On the contrary, S1P2 was found to negatively regulate cell migration. Collectively, these results are in favour of an important function of S1P in satellite cell biology that could in principle be exploited as novel pharmacological target for improving skeletal muscle regeneration.


Biochemical Journal | 2002

Sphingosine 1-phosphate evokes calcium signals in C2C12 myoblasts via Edg3 and Edg5 receptors.

Elisabetta Meacci; Francesca Cencetti; Lucia Formigli; Roberta Squecco; Chiara Donati; Bruno Tiribilli; Franco Quercioli; Sandra Zecchi Orlandini; Fabio Francini; Paola Bruni

Sphingosine 1-phosphate (SPP) is a bioactive lipid that exerts multiple biological effects in a large variety of cell types, acting as either an intracellular messenger or an extracellular ligand coupled to Edg-family receptors (where Edg stands for endothelial differentiation gene). Here we report that in C(2)C(12) myoblasts SPP elicited significant Ca(2+) mobilization. Analysis of the process using a confocal laser-scanning microscope showed that the Ca(2+) response occurred in a high percentage of cells, despite variations in amplitude and kinetics. Quantitative analysis of SPP-induced Ca(2+) transients performed with a spectrophotofluorimeter showed that the rise in Ca(2+) was strictly dependent on availability of extracellular Ca(2+). Cell treatment with pertussis toxin partially prevented the Ca(2+) response induced by SPP, indicating that G(i)-coupled-receptors were involved. Indeed, SPP action was shown to be mediated by agonist-specific Edg receptors. In particular, suramin, an antagonist of the SPP-specific receptor Edg3, as well as down-regulation of Edg3 by cell transfection with antisense oligodeoxyribonucleotides (ODN), significantly reduced agonist-mediated Ca(2+) mobilization. Moreover, an antisense ODN designed to inhibit Edg5 expression also decreased the SPP-induced rise in Ca(2+), although to a lesser extent than that observed by inhibiting Edg3. On the contrary, the SPP response was unaffected in myoblasts loaded with antisense ODN specific for Edg1. Remarkably, the concomitant inhibition of Edg3 and Edg5 receptors abolished the SPP-induced Ca(2+) increase, supporting the notion that Ca(2+) mobilization in C(2)C(12) cells induced by SPP is a receptor-mediated process that involves Edg3 and Edg5, but not Edg1.


FEBS Letters | 2000

Permissive role of protein kinase Cα but not protein kinase Cδ in sphingosine 1‐phosphate‐induced RhoA activation in C2C12 myoblasts

Elisabetta Meacci; Chiara Donati; Francesca Cencetti; Elena Romiti; Paola Bruni

Rho GTPases participate in various important signaling pathways and have been implicated in myogenic differentiation. Here the first evidence is provided that in C2C12 myoblasts sphingosine 1‐phosphate (SPP) rapidly and transiently induced membrane association of RhoA in a pertussis toxin‐insensitive manner. The bioactive lipid preferentially relocalized the GTPase to Golgi‐enriched membrane. Translocation of RhoA was abolished by inhibition or down‐regulation of protein kinase C (PKC). Notably, treatment with Gö6976, an inhibitor of conventional PKCs, which selectively blocked PKCα in these cells, prevented SPP‐induced RhoA translocation. Conversely rottlerin, a selective inhibitor of PKCδ, was without effect, demonstrating that SPP signaling to RhoA involves PKCα but not PKCδ activation. This novel functional relationship between the two proteins may have a role in SPP‐mediated regulation of downstream effectors.


FEBS Letters | 2007

Tumor necrosis factor‐α exerts pro‐myogenic action in C2C12 myoblasts via sphingosine kinase/S1P2 signaling

Chiara Donati; Paola Nincheri; Francesca Cencetti; Elena Rapizzi; Marta Farnararo; Paola Bruni

In this study, we report that low doses of tumor necrosis factor‐α (TNFα) promote myogenesis in C2C12 myoblasts. Moreover, the cytokine increased sphingosine kinase (SphK) activity and induced SphK1 translocation to membranes. The inhibition of SphK functionality by various approaches abrogated the pro‐myogenic effect of TNFα. Moreover, silencing of S1P2 impaired the positive action of TNFα on myogenesis. These results represent the first evidence that SphK/S1P2 axis is required for the regulation of myogenesis by TNFα. In view of the physiological role of TNFα in muscle regeneration, the present finding reinforces the notion that SphK/S1P2 signaling is critically implicated in myogenesis.


Biochimica et Biophysica Acta | 2003

Down-regulation of EDG5/S1P2 during myogenic differentiation results in the specific uncoupling of sphingosine 1-phosphate signalling to phospholipase D

Elisabetta Meacci; Francesca Cencetti; Chiara Donati; Francesca Nuti; Marta Farnararo; Takayuki Kohno; Yasuyuki Igarashi; Paola Bruni

The bioactive lipid sphingosine 1-phosphate (S1P) is known to exert powerful biological effects through the interaction with various members of the endothelial differentiation gene (EDG) receptor family, recently renamed S1P receptors. In the present study, evidence is provided that differentiation of C2C12 myoblasts into myotubes was accompanied by profound changes of EDG/S1P receptor expression. Indeed, in differentiated cells a significant increase of EDG3/S1P3 together with a large decrease of EDG5/S1P2 expression at mRNA as well as protein level was detected. Moreover, S1P was capable to initiate the signalling pathways downstream to cytosolic Ca(2+) increase in myotubes, similarly to that observed in myoblasts, whereas the signalling of the bioactive lipid to phospholipase D (PLD), but not that of bradykinin (BK) or lysophosphatidic acid (LPA), was found impaired in differentiated cells. Intriguingly, overexpression of EDG5/S1P2, but not EDG1/S1P1 or EDG3/S1P3, potentiated the efficacy of S1P to stimulate PLD, strongly suggesting a role for EDG5/S1P2 in the signalling to PLD. This view was also supported by the marked reduction of S1P-induced PLD activity in myoblasts loaded with antisense oligodeoxyribonucleotides (ODN) to EDG5/S1P2. Furthermore, overexpression of EDG5/S1P2 rescued the coupling of S1P signalling to PLD in C2C12 myotubes. Experimental evidence here provided supports the notion that EDG5/S1P2 plays a dominant role in the coupling of S1P to PLD in myoblasts and that the down-regulation of the receptor subtype is responsible for the specific uncoupling of S1P signalling to PLD in myotubes.


Molecular and Cellular Biochemistry | 2008

Sphingosine 1-phosphate differentially regulates proliferation of C2C12 reserve cells and myoblasts

Elena Rapizzi; Chiara Donati; Francesca Cencetti; Paola Nincheri; Paola Bruni

The effect of sphingosine 1-phosphate (S1P) on the proliferative response to low serum was examined in two closely related cell populations, such as C2C12 reserve cells and myoblasts. S1P reduced DNA synthesis promoted by serum in myoblasts, whereas it enhanced the mitogenic response to serum in reserve cells. By employing selective S1P receptor agonist and antagonists, the co-mitogenic action of S1P in reserve cells was shown to depend mainly on S1P1. Real time PCR analysis revealed distinct S1P receptor pattern expression, which however could not account for the differential action of S1P in C2C12 reserve cells and myoblasts, thereby suggesting that the cell-specific responsiveness to S1P may depend on a different functional coupling of S1P1. This study discloses a unique pleiotropic effect of S1P which stimulates proliferation of muscle resident stem cells, such as reserve cells, and favours the growth arrest of committed progenitors cells, such as myoblasts, required for their subsequent myogenic differentiation.

Collaboration


Dive into the Francesca Cencetti's collaboration.

Top Co-Authors

Avatar

Paola Bruni

University of Florence

View shared research outputs
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