Network


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

Hotspot


Dive into the research topics where Chiara Ruzza is active.

Publication


Featured researches published by Chiara Ruzza.


British Journal of Pharmacology | 2009

Neuropeptide S is a stimulatory anxiolytic agent: a behavioural study in mice

Anna Rizzi; Raffaella Vergura; Giuliano Marzola; Chiara Ruzza; Remo Guerrini; Severo Salvadori; Domenico Regoli; Girolamo Calo

Neuropeptide S (NPS) was recently identified as the endogenous ligand of an orphan receptor, now referred to as the NPS receptor. In vivo, NPS produces a unique behavioural profile by increasing wakefulness and exerting anxiolytic‐like effects. In the present study, we further evaluated the effects of in vivo supraspinal NPS in mice.


Journal of Pharmacology and Experimental Therapeutics | 2009

In Vitro and in Vivo Pharmacological Characterization of the Neuropeptide S Receptor Antagonist [d-Cys(tBu)5]Neuropeptide S

Valeria Camarda; Anna Rizzi; Chiara Ruzza; Silvia Zucchini; Giuliano Marzola; Erika Marzola; Remo Guerrini; Severo Salvadori; Rainer K. Reinscheid; Domenico Regoli; Girolamo Calo

Neuropeptide S (NPS) was identified as the endogenous ligand of an orphan receptor now referred to as the NPS receptor (NPSR). In the frame of a structure-activity study performed on NPS Gly5, the NPSR ligand [d-Cys(tBu)5]NPS was identified. [d-Cys(tBu)5]NPS up to 100 μM did not stimulate calcium mobilization in human embryonic kidney (HEK) 293 cells stably expressing the mouse NPSR; however, in a concentration-dependent manner, the peptide inhibited the stimulatory effects elicited by 10 and 100 nM NPS (pKB, 6.62). In Schild analysis experiments [d-Cys(tBu)5]NPS (0.1–100 μM) produced a concentration-dependent and parallel rightward shift of the concentration-response curve to NPS, showing a pA2 value of 6.44. Ten micromolar [d-Cys(tBu)5]NPS did not affect signaling at seven NPSR unrelated G-protein-coupled receptors. In the mouse righting reflex (RR) recovery test, NPS given at 0.1 nmol i.c.v. reduced the percentage of animals losing the RR in response to 15 mg/kg diazepam and their sleeping time. [d-Cys(tBu)5]NPS (1–10 nmol) was inactive per se but dose-dependently antagonized the arousal-promoting action of NPS. Finally, NPSR-deficient mice were similarly sensitive to the hypnotic effects of diazepam as their wild-type littermates. However, the arousal-promoting action of 1 nmol NPS could be detected in wild-type but not in mutant mice. In conclusion, [d-Cys(tBu)5]NPS behaves both in vitro and in vivo as a pure and selective NPSR antagonist but with moderate potency. Moreover, using this tool together with receptor knockout mice studies, we demonstrated that the arousal-promoting action of NPS is because of the selective activation of the NPSR protein.


Journal of Medicinal Chemistry | 2009

Synthesis and biological activity of human neuropeptide S analogues modified in position 5: identification of potent and pure neuropeptide S receptor antagonists.

Remo Guerrini; Valeria Camarda; Claudio Trapella; Girolamo Calo; Anna Rizzi; Chiara Ruzza; Stella Fiorini; Erika Marzola; Rainer K. Reinscheid; Domenico Regoli; Severo Salvadori

Neuropeptide S (NPS), the endogenous ligand of a previously orphan receptor now named NPSR, regulates various biological functions in the brain, including arousal, locomotion, anxiety, and food intake. Here we report on a focused structure-activity study of Gly5, which has been replaced with L and D amino acids. Fifteen NPS related peptides were synthesized and pharmacologically tested for intracellular calcium mobilization using HEK293 cells stably expressing the mouse NPSR. The results of this study demonstrated that peptide potency is inversely related to the side chain size, while peptide efficacy strongly depends on the relative L and D configuration, with the L amino acids favoring agonist while D amino acids display antagonist pharmacological activity. [D-Val5]NPS behaved as NPSR pure antagonist in HEK293(mNPSR) cells showing the highest potency (pK(B) 7.56) among this series of peptides. The antagonist action of [D-Val5]NPS was confirmed in vivo in mice, where the peptide at a dose of 10 nmol completely blocked the stimulatory effect of 0.1 nmol NPS on locomotor activity.


Peptides | 2010

Further studies on the pharmacological profile of the neuropeptide S receptor antagonist SHA 68.

Chiara Ruzza; Anna Rizzi; Claudio Trapella; Michela Pelà; Valeria Camarda; Valentina Ruggieri; Monica Filaferro; Carlo Cifani; Rainer K. Reinscheid; Giovanni Vitale; Roberto Ciccocioppo; Severo Salvadori; Remo Guerrini; Girolamo Calo

Neuropeptide S (NPS) regulates various biological functions by selectively activating the NPS receptor (NPSR). Previous studies demonstrated that the non-peptide molecule SHA 68 acts as a selective NPSR antagonist. In the present study the pharmacological profile of SHA 68 has been further investigated in vitro and in vivo. In cells expressing the mouse NPSR SHA 68 was inactive per se up to 10microM while it antagonized NPS-stimulated calcium mobilization in a competitive manner showing a pA(2) value of 8.06. In the 10-50mg/kg range of doses, SHA 68 counteracted the stimulant effects elicited by NPS, but not those of caffeine, in mouse locomotor activity experiments. In the mouse righting reflex assay SHA 68 fully prevented the arousal-promoting action of the peptide. The anxiolytic-like effects of NPS were slightly reduced by SHA 68 in the mouse open field, fully prevented in the rat elevated plus maze and partially antagonized in the rat defensive burying paradigm. Finally, SHA 68 was found poorly active in antagonizing the NPS inhibitory effect on palatable food intake in rats. In all assays SHA 68 did not produce any effect per se. In conclusion, the present study demonstrated that SHA 68 behaves as a selective NPSR antagonist that can be used to characterize the in vivo actions of NPS. However the usefulness of this research tool is limited by its poor pharmacokinetic properties.


Neuropharmacology | 2012

Behavioural phenotypic characterization of CD-1 mice lacking the neuropeptide S receptor

Chiara Ruzza; Alice Pulga; Anna Rizzi; Giuliano Marzola; Remo Guerrini; Girolamo Calo

Neuropeptide S (NPS) is the endogenous ligand of a previously orphan receptor now named NPSR. In the brain NPS regulates several biological functions including anxiety, arousal, locomotion, food intake, learning and memory, pain and drug abuse. Mice lacking the NPSR gene (NPSR(-/-)) represent an useful tool to investigate the neurobiology of the NPS/NPSR system. NPSR(-/-) mice have been generated in a 129S6/SvEv genetic background. In the present study we generated CD-1 congenic NPSR(+/+) and NPSR(-/-) mice and investigated their phenotype and sensitivity to NPS in various behavioural assays. The phenotype analysis revealed no locomotor differences between NPSR(+/+) and NPSR(-/-) mice. The behaviour of NPSR(+/+) and NPSR(-/-) mice in the righting reflex test was superimposable. No differences were recorded between the two genotypes in the elevated plus maze, open field and stress-induced hyperthermia tests, with the exception of rearing behaviour that was reduced in knockout animals. Moreover the behaviour of NPSR(+/+) and NPSR(-/-) mice in the forced swimming, novel object recognition and formalin assays was similar. The stimulatory effects of NPS in the locomotor activity test and its anxiolytic-like actions in the elevated plus maze and open field assays were evident in NPSR(+/+) but not NPSR(-/-) animals. In conclusion, the present study indicates that the NPS/NPSR system does not tonically control locomotion, sensitivity to diazepam, anxiety, depressive-like behaviours, memory and pain transmission in mice. Furthermore our results clearly show that the product of the NPSR gene represents the mandatory protein for all the NPS biological effects so far described.


European Journal of Neuroscience | 2012

Anxiolytic- and panicolytic-like effects of Neuropeptide S in the mouse elevated T-maze

Alice Pulga; Chiara Ruzza; Anna Rizzi; Remo Guerrini; Girolamo Calo

Neuropeptide S (NPS) regulates various biological functions by selectively activating the NPS receptor (NPSR). Recently, epidemiological studies revealed an association between NPSR single nucleotide polymorphisms and susceptibility to panic disorders. Here we investigated the effects of NPS in mice subjected to the elevated T maze (ETM), an assay which has been proposed to model anxiety and panic. Diazepam [1 mg/kg, intraperitoneally (i.p.)] elicited clear anxiolytic effects reducing the latency to emerge from the closed to the open (CO) arm without modifying the latencies from the open to the closed (OC) arm. By contrast, chronic fluoxetine (10 mg/kg i.p., once a day for 21 days) selectively increased OC latency, suggesting a panicolytic‐like effect. NPS given intracerebroventricularly at 0.001–1 nmol elicited both anxiolytic‐ and panicolytic‐like effects. However, although the NPS anxiolytic dose–response curve displayed the classical sigmoidal shape, the dose–response curve of the putative panicolytic‐like effect was bell shaped with peak effect at 0.01 nmol. The behaviour of wild‐type [NPSR(+/+)] and receptor knock out [NPSR(−/−)] mice in the ETM task was superimposable. NPS at 0.01 nmol elicited anxiolytic‐ and panicolytic‐like effects in NPSR(+/+) but not in NPSR(−/−) mice. In conclusion, this study demonstrated that NPS, via selective activation of the NPSR, promotes both anxiolytic‐ and panicolytic‐like actions in the mouse ETM.


Journal of Medicinal Chemistry | 2009

Further Studies at Neuropeptide S Position 5: Discovery of Novel Neuropeptide S Receptor Antagonists

Remo Guerrini; Valeria Camarda; Claudio Trapella; Girolamo Calo; Anna Rizzi; Chiara Ruzza; Stella Fiorini; Erika Marzola; Rainer K. Reinscheid; Domenico Regoli; Severo Salvadori

Neuropeptide S (NPS) regulates various biological functions by activating the NPS receptor (NPSR). Previous studies demonstrated that the substitution of Gly(5) with d-amino acids generates NPSR antagonists. Eleven [d-Xaa(5)]NPS derivatives were synthesized and pharmacologically tested measuring [Ca(2+)](i) in HEK293(mNPSR) cells. The results confirmed that the [d-Xaa(5)] substitution promotes antagonist activity with potency inversely related to the side chain size and allowed identification of the novel potent NPSR peptide antagonist [(t)Bu-d-Gly(5)]NPS.


British Journal of Pharmacology | 2014

In vitro and in vivo pharmacological characterization of nociceptin/orphanin FQ tetrabranched derivatives

Anna Rizzi; Davide Malfacini; Maria Camilla Cerlesi; Chiara Ruzza; Erika Marzola; Mark F. Bird; David J. Rowbotham; Severo Salvadori; Remo Guerrini; David G. Lambert; Girolamo Calo

An innovative chemical approach, named peptide welding technology (PWT), allows the synthesis of multibranched peptides with extraordinary high yield, purity and reproducibility. With this approach, three different tetrabranched derivatives of nociceptin/orphanin FQ (N/OFQ) have been synthesized and named PWT1‐N/OFQ, PWT2‐N/OFQ and PWT3‐N/OFQ. In the present study we investigated the in vitro and in vivo pharmacological profile of PWT N/OFQ derivatives and compared their actions with those of the naturally occurring peptide.


Journal of Medicinal Chemistry | 2011

Synthesis and Separation of the Enantiomers of the Neuropeptide S Receptor Antagonist (9R/S)-3-Oxo-1,1-diphenyl-tetrahydro-oxazolo[3,4-a]pyrazine-7-carboxylic Acid 4-Fluoro-benzylamide (SHA 68)

Claudio Trapella; Michela Pelà; Luisa Del Zoppo; Girolamo Calo; Valeria Camarda; Chiara Ruzza; Alberto Cavazzini; Valentina Costa; Valerio Bertolasi; Rainer K. Reinscheid; Severo Salvadori; Remo Guerrini

This study reports the synthesis, chromatographic separation, and pharmacological evaluation of the two enantiomers of the neuropeptide S receptor (NPSR) antagonist (9R/S)-3-oxo-1,1-diphenyl-tetrahydro-oxazolo[3,4-a]pyrazine-7-carboxylic acid 4-fluoro-benzylamide (SHA 68). The (9R)-3-oxo-1,1-diphenyl-tetrahydro-oxazolo[3,4-a]pyrazine-7-carboxylic acid 4-fluoro-benzylamide (compound 10) and (9S)-3-oxo-1,1-diphenyl-tetrahydro-oxazolo[3,4-a]pyrazine-7-carboxylic acid 4-fluoro-benzylamide (compound 10a) were synthesized and their purity assessed by chiral chromatography. The absolute configuration of the enantiomer 10 has been assigned from the crystal structure of the corresponding (S)-phenyl ethyl amine derivative 8. Calcium mobilization studies performed on cells expressing the recombinant NPSR demonstrated that compound 10 is the active enantiomer while the contribution of 10a to the NPSR antagonist properties of the racemic mixture is negligible.


Pharmacology Research & Perspectives | 2016

Pharmacological characterization of cebranopadol a novel analgesic acting as mixed nociceptin/orphanin FQ and opioid receptor agonist

Anna Rizzi; Maria Camilla Cerlesi; Chiara Ruzza; Davide Malfacini; Federica Ferrari; Sara Bianco; Tommaso Costa; Remo Guerrini; Claudio Trapella; Girolamo Calo

The aim of the study was to investigate the in vitro and in vivo pharmacological profile of cebranopadol, a novel agonist for opioid and nociceptin/orphanin FQ (N/OFQ) receptors (NOP). In vitro cebranopadol was assayed in calcium mobilization studies in cells coexpressing NOP or opioid receptors and chimeric G‐proteins and in a bioluminescence resonance energy transfer (BRET) assay for studying receptor interaction with G‐protein and β‐arrestin 2. The mouse tail withdrawal and formalin tests were used for investigating cebranopadol antinociceptive properties. In calcium mobilization studies cebranopadol showed the following rank order of potency NOP = mu > kappa ≥ delta. In BRET studies, cebranopadol promoted NOP and mu receptors interaction with G‐protein with similar high potency and efficacy. However, cebranopadol did not stimulated NOP–β‐arrestin 2 interactions and displayed reduced potency at mu/β‐arrestin 2. In vivo, cebranopadol exhibits highly potent and extremely long‐lasting antinociceptive effects. The effects of cebranopadol in the tail withdrawal assay were sensitive to both SB‐612111 and naloxone. Collectively the present results confirm and extend previous finding demonstrating that cebranopadol, by acting as mixed NOP/opioid receptor agonist, elicits robust analgesic effects in different pain models.

Collaboration


Dive into the Chiara Ruzza'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

Elaine C. Gavioli

Federal University of Rio Grande do Norte

View shared research outputs
Researchain Logo
Decentralizing Knowledge