Network


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

Hotspot


Dive into the research topics where Arianna Carolina Rosa is active.

Publication


Featured researches published by Arianna Carolina Rosa.


Neurochemistry International | 2009

PPARγ stimulation promotes mitochondrial biogenesis and prevents glucose deprivation-induced neuronal cell loss

Gianluca Miglio; Arianna Carolina Rosa; Lorenza Rattazzi; Massimo Collino; Grazia Lombardi; Roberto Fantozzi

Peroxisome proliferator-activated receptor (PPAR)gamma stimulation provides protection in several models of neurological disorders, but the mechanisms underlying these effects remain to be fully elucidated. Here we have studied whether two PPARgamma agonists, pioglitazone and rosiglitazone, prevent loss of differentiated SH-SY5Y cells transiently exposed to glucose deprivation (GD). Nanomolar drug concentrations prevented GD-induced cell loss in a concentration- and time-dependent manner. These effects were abolished by malonate, a reversible mitochondrial Complex II inhibitor, while significantly potentiated by pyruvate, thus suggesting that they are related to mitochondrial function. During cell pretreatment, PPARgamma agonists promoted biogenesis of functional mitochondria, as indicated by the up-regulation of PPARgamma coactivator (PGC)-1alpha, NRF1, TFAM, cytochrome c oxidase subunit (CO) I and CO IV, and the increased level of mtDNA, while did not significantly change mitochondrial membrane potential. In addition, the analysis of the concentration-response and time-course curves for the protective effects and the up-regulation of mitochondrial biogenesis markers suggests that mitochondrial biogenesis and cell loss prevention are related effects. In conclusion our data indicate that a prolonged PPARgamma stimulation, by repeated administration of nanomolar pioglitazone or rosiglitazone concentrations, decreases GD-induced loss of differentiated SH-SY5Y cells. In addition, they suggest that mitochondrial biogenesis may contribute to these effects.


British Journal of Pharmacology | 2013

The role of histamine in neurogenic inflammation

Arianna Carolina Rosa; Roberto Fantozzi

The term ‘neurogenic inflammation’ has been adopted to describe the local release of inflammatory mediators, such as substance P and calcitonin gene‐related peptide, from neurons. Once released, these neuropeptides induce the release of histamine from adjacent mast cells. In turn, histamine evokes the release of substance P and calcitonin gene‐related peptide; thus, a bidirectional link between histamine and neuropeptides in neurogenic inflammation is established. The aim of this review is to summarize the most recent findings on the role of histamine in neurogenic inflammation, with particular regard to nociceptive pain, as well as neurogenic inflammation in the skin, airways and bladder.


Journal of Cellular and Molecular Medicine | 2013

Acute treatment with relaxin protects the kidney against ischaemia/reperfusion injury

Massimo Collino; Mara Rogazzo; Alessandro Pini; Elisa Benetti; Arianna Carolina Rosa; Fausto Chiazza; Roberto Fantozzi; Daniele Bani; Emanuela Masini

Although recent preclinical and clinical studies have demonstrated that recombinant human relaxin (rhRLX) may have important therapeutic potential in acute heart failure and chronic kidney diseases, the effects of acute rhRLX administration against renal ischaemia/reperfusion (I/R) injury have never been investigated. Using a rat model of 1‐hr bilateral renal artery occlusion followed by 6‐hr reperfusion, we investigated the effects of rhRLX (5 μg/Kg i.v.) given both at the beginning and after 3 hrs of reperfusion. Acute rhRLX administration attenuated the functional renal injury (increase in serum urea and creatinine), glomerular dysfunction (decrease in creatinine clearance) and tubular dysfunction (increase in urinary excretion of N‐acetyl‐β‐glucosaminidase) evoked by renal I/R. These beneficial effects were accompanied by a significant reduction in local lipid peroxidation, free radical‐induced DNA damage and increase in the expression/activity of the endogenous antioxidant enzymes Mn‐ and CuZn‐superoxide dismutases (SOD). Furthermore, rhRLX administration attenuated the increase in leucocyte activation, as suggested by inhibition of myeloperoxidase activity, intercellular‐adhesion‐molecule‐1 expression, interleukin (IL)‐1β, IL‐18 and tumour necrosis factor‐α production as well as increase in IL‐10 production. Interestingly, the reduced oxidative stress status and neutrophil activation here reported were associated with rhRLX‐induced activation of endothelial nitric oxide synthase and up‐regulation of inducible nitric oxide synthase, possibly secondary to activation of Akt and the extracellular signal‐regulated protein kinase (ERK) 1/2, respectively. Thus, we report herein that rhRLX protects the kidney against I/R injury by a mechanism that involves changes in nitric oxide signalling pathway.


British Journal of Pharmacology | 2009

Celecoxib decreases expression of the adhesion molecules ICAM‐1 and VCAM‐1 in a colon cancer cell line (HT29)

Margherita Gallicchio; Arianna Carolina Rosa; Chiara Dianzani; L Brucato; Elisa Benetti; Massimo Collino; Roberto Fantozzi

We investigated the ability of celecoxib, a selective cyclooxygenase‐2 (COX‐2) inhibitor, to modulate expression of ICAM‐1 and VCAM‐1 in the colon cancer cell line HT29.


Free Radical Biology and Medicine | 2011

Peroxisome proliferator-activated receptor β/δ agonism protects the kidney against ischemia/reperfusion injury in diabetic rats

Massimo Collino; Elisa Benetti; Gianluca Miglio; Sara Castiglia; Arianna Carolina Rosa; Manuela Aragno; Christoph Thiemermann; Roberto Fantozzi

Diabetes is an important risk factor for ischemic acute kidney injury, whose pharmacological treatment remains an unmet medical need. The peroxisome proliferator-activated receptor (PPAR) β/δ is highly expressed in the kidney, although its role has not yet been elucidated. Here, we used an in vivo model of renal ischemia/reperfusion (I/R) in streptozotocin-induced diabetic rats (i) to evaluate whether diabetes increases kidney susceptibility to I/R injury and (ii) to investigate the effects of PPARβ/δ activation. The degree of renal injury (1h ischemia/6h reperfusion) was significantly increased in diabetic rats compared with nondiabetic littermates. PPARβ/δ expression was increased after I/R, with the highest levels in diabetic rats. Administration of the selective PPARβ/δ agonist GW0742 attenuated the renal dysfunction, leukocyte infiltration, and formation of interleukin-6 and tumor necrosis factor-α. These effects were accompanied by an increased expression of the suppressor of cytokine signaling (SOCS)-3, which plays a critical role in the cytokine-activated signaling pathway. The beneficial effects of GW0742 were attenuated by the selective PPARβ/δ antagonist GSK0660. Thus, we report herein that PPARβ/δ activation protects the diabetic kidney against I/R injury by a mechanism that may involve changes in renal expression of SOCS-3 resulting in a reduced local inflammatory response.


British Journal of Pharmacology | 2009

Substance P-induced cyclooxygenase-2 expression in human umbilical vein endothelial cells

Margherita Gallicchio; Arianna Carolina Rosa; Elisa Benetti; Massimo Collino; Chiara Dianzani; Roberto Fantozzi

Substance P (SP) is a neuropeptide involved in neurogenic inflammation and an agonist for NK1, NK2, and NK3 receptors. SP induces prostaglandin (PG) production in various cell types, and these eicosanoids are responsible for numerous inflammatory and vascular effects. Cyclooxygenase (COX) are needed to convert arachidonic acid to PGs. The study evaluated the effect of SP on COX expression in human umbilical vein endothelial cells (HUVEC). COX‐2 protein expression was upregulated by SP with a peak at 100 nM and at 20 h; in the same experimental conditions COX‐1 protein expression was unchanged. A correlation between COX‐2 expression and PGI2 and PGE2 release was detected. Dexamethasone (DEX) inhibited SP‐mediated COX‐2 expression. Mitogen‐activated protein kinases (MAPK) p38 and p42/44 were activated by SP, whereas SB202190 and PD98059, inhibitors of these kinases, blocked COX‐2 expression. 5,5‐dimethyl‐3‐(3‐fluorophenyl)‐4‐(4‐methylsulphonyl)phenyl‐2(5H)‐furanone (DFU), an experimental selective COX‐2 inhibitor, blocked SP‐induced PG release. By RT–PCR and Western blot analysis, we demonstrated that NK1 and NK2 but not NK3 receptors are present on HUVEC. Selective NK1 and NK2 agonists, namely [Sar9, Met(O2)11]SP and [β‐Ala8] NKA(4–10), upregulated COX‐2 protein expression and PG production, whereas senktide (Suc–Asp–Phe–MePhe–Gly–Leu–Met–NH2), a selective NK3 agonist, was ineffective in this respect. The NK1 selective antagonist L703,606 ((cis)‐2‐(diphenylmethyl)‐N‐((2‐iodophenyl)‐methyl)‐1‐azabicyclo(2.2.2)octan‐3‐amine) and the NK2 selective antagonist SR 48,968 ((S)‐N‐methyl‐N‐(4‐(4‐acetylamino‐4‐phenylpiperidino)‐2‐(3,4 dichlorophenyl)butyl) benzamide) competitively antagonised SP‐induced effects. The study shows HUVEC to possess functional NK1 and NK2 receptors, which mediate the ability of SP to induce expression of COX‐2 in HUVEC, thus showing a previously‐undetected effect of SP on endothelial cells.


British Journal of Pharmacology | 2011

The subtypes of peroxisome proliferator‐activated receptors expressed by human podocytes and their role in decreasing podocyte injury

Gianluca Miglio; Arianna Carolina Rosa; Lorenza Rattazzi; Cristina Grange; Massimo Collino; Giovanni Camussi; Roberto Fantozzi

BACKGROUND AND PURPOSE Peroxisome proliferator‐activated receptors (PPARs) are ligand‐activated transcription factors, and three subtypes (α, β and γ) have been identified. PPAR activation has been reported to decrease renal injury and markers of glomerular dysfunction in models of renal ischemia/reperfusion (I/R). However, both the I/R effects and the effects of PPAR agonists on podocytes, an integral cellular part of the glomerular filtration barrier, remain to be established.


Neuroscience Letters | 2009

PPARγ stimulation promotes neurite outgrowth in SH-SY5Y human neuroblastoma cells

Gianluca Miglio; Lorenza Rattazzi; Arianna Carolina Rosa; Roberto Fantozzi

Several evidences indicate that PPARgamma stimulation promotes neuronal differentiation. However, to date, no data describe the effects of PPARgamma agonists on neurite outgrowth. Here we have evaluated the effects of pioglitazone, a synthetic PPARgamma agonist, on differentiation and neurite outgrowth in SH-SY5Y human neuroblastoma cells. Our results show that pioglitazone promotes cell differentiation and the outgrowth of cell processes in a concentration-dependent manner with the maximal effect at 100 nM-1 microM. It significantly increases both the mean process length and the percentage of neurite-bearing cells. In addition, these effects are accompanied by significant activation of p42 and p44 mitogen-activated protein kinases. In conclusion, albeit preliminary, these findings suggest the possibility that PPARgamma stimulation may contribute to the development and maintenance of a proper neuronal connectivity within neuronal networks.


British Journal of Pharmacology | 2009

Celecoxib modulates adhesion of HT29 colon cancer cells to vascular endothelial cells by inhibiting ICAM-1 and VCAM-1 expression

Chiara Dianzani; L Brucato; Margherita Gallicchio; Arianna Carolina Rosa; Massimo Collino; Roberto Fantozzi

Cyclooxygenase‐2 (COX‐2) is highly expressed during inflammation and can promote the progression of colorectal cancer. Interactions between cancer cells and vascular endothelial cells are key events in this process. Recently, the selective COX‐2 inhibitor, celecoxib, was shown to inhibit expression of the adhesion molecules, ICAM‐1 and VCAM‐1, in the human colon cancer cell line HT29 and to inhibit adhesion of HT29 cells to FCS‐coated plastic wells. Here, we evaluated the effects of celecoxib on adhesion of HT29 cells to human umbilical vein endothelial cells (HUVEC), mediated by ICAM‐1 and VCAM‐1, to assess further the potential protective effects of celecoxib on cancer development.


British Journal of Pharmacology | 2012

Cholesteryl butyrate solid lipid nanoparticles inhibit the adhesion and migration of colon cancer cells

Rosalba Minelli; L. Serpe; Piergiorgio Pettazzoni; Valerio Giacomo Minero; Giuseppina Barrera; Casimiro Luca Gigliotti; Riccardo Mesturini; Arianna Carolina Rosa; P. Gasco; N. Vivenza; Elisabetta Muntoni; Roberto Fantozzi; Umberto Dianzani; Gian Paolo Zara; Chiara Dianzani

BACKGROUND AND PURPOSE Cholesteryl butyrate solid lipid nanoparticles (cholbut SLN) provide a delivery system for the anti‐cancer drug butyrate. These SLN inhibit the adhesion of polymorphonuclear cells to the endothelium and may act as anti‐inflammatory agents. As cancer cell adhesion to endothelium is crucial for metastasis dissemination, here we have evaluated the effect of cholbut SLN on adhesion and migration of cancer cells.

Collaboration


Dive into the Arianna Carolina Rosa'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