Network


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

Hotspot


Dive into the research topics where Davide Malfacini is active.

Publication


Featured researches published by Davide Malfacini.


Structure | 2015

The Importance of Ligand-Receptor Conformational Pairs in Stabilization: Spotlight on the N/OFQ G Protein-Coupled Receptor

Rebecca L. Miller; Aaron A. Thompson; Claudio Trapella; Remo Guerrini; Davide Malfacini; Nilkanth Patel; Gye Won Han; Vadim Cherezov; Girolamo Calo; Vsevolod Katritch; Raymond C. Stevens

Understanding the mechanism by which ligands affect receptor conformational equilibria is key in accelerating membrane protein structural biology. In the case of G protein-coupled receptors (GPCRs), we currently pursue a brute-force approach for identifying ligands that stabilize receptors and facilitate crystallogenesis. The nociceptin/orphanin FQ peptide receptor (NOP) is a member of the opioid receptor subfamily of GPCRs for which many structurally diverse ligands are available for screening. We observed that antagonist potency is correlated with a ligands ability to induce receptor stability (Tm) and crystallogenesis. Using this screening strategy, we solved two structures of NOP in complex with top candidate ligands SB-612111 and C-35. Docking studies indicate that while potent, stabilizing antagonists strongly favor a single binding orientation, less potent ligands can adopt multiple binding modes, contributing to their low Tm values. These results suggest a mechanism for ligand-aided crystallogenesis whereby potent antagonists stabilize a single ligand-receptor conformational pair.


British Journal of Pharmacology | 2013

Acute and chronic antiparkinsonian effects of the novel nociceptin/orphanin FQ receptor antagonist NiK-21273 in comparison with SB-612111

Matteo Marti; Flora Mela; Mirco Budri; Mattia Volta; Davide Malfacini; Stefano Molinari; Nurulain T. Zaveri; S Ronzoni; P Petrillo; Girolamo Calo; Michele Morari

Nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor antagonists have been proposed as a novel therapeutic approach to Parkinsons disease. Main limitations of previous studies were the use of structurally similar compounds and the evaluation of their acute effects only. We report here on the acute and long‐term antiparkinsonian effects of the novel compound 2‐[3‐[4‐(2‐chloro‐6‐fluoro‐phenyl)‐piperidin‐1‐ylmethyl]‐2‐(morpholine‐4‐carbonyl)‐indol‐1‐yl]‐acetamide (NiK‐21273) in comparison with the potent and selective NOP receptor antagonist SB‐612111.


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.


Bioorganic & Medicinal Chemistry | 2014

A novel and facile synthesis of tetra branched derivatives of nociceptin/orphanin FQ.

Remo Guerrini; Erika Marzola; Claudio Trapella; Michela Pelà; Stefano Molinari; Maria Camilla Cerlesi; Davide Malfacini; Anna Rizzi; Severo Salvadori; Girolamo Calo

Branched peptides have been found to be useful in several research fields however their synthesis and purification is complicated. Here we present a novel and facile synthesis of tetra branched derivatives of nociceptin/orphanin FQ (N/OFQ). Three N/OFQ tetra branched derivatives were prepared using novel cores (PWT1, PWT2 and PWT3) containing a maleimido moiety. [Cys(18)]N/OFQ-NH2 was linked to the cores via thiol-Michael reaction characterized by high yield and purity of the desired final product. In the electrically stimulated mouse vas deferens PWT-N/OFQ derivatives mimicked the inhibitory action of the natural sequence showing similar maximal effects and 3 fold higher potencies. The NOP selective antagonist SB-612111 antagonized the effects of N/OFQ and PWT derivatives with similar pKB values (8.02-8.48). In vivo after supraspinal administration PWT2-N/OFQ stimulated food intake in mice mimicking the action of N/OFQ. Compared to the natural peptide PWT2-N/OFQ was 40 fold more potent and elicited larger effects. These findings suggest that the PWT chemical strategy can be successfully applied to biologically active peptides to generate, with unprecedented high purity and yield, tetra branched derivatives displaying an in vitro pharmacological profile similar to that of the natural sequence associated, in vivo, to increased potency and effectiveness.


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.


PLOS ONE | 2015

Pharmacological Profile of Nociceptin/Orphanin FQ Receptors Interacting with G-Proteins and β-Arrestins 2

Davide Malfacini; Caterina Ambrosio; M. C. Gro; M. Sbraccia; Claudio Trapella; Remo Guerrini; Massimo Bonora; Paolo Pinton; Tommaso Costa; Girolamo Calo

Nociceptin/orphanin FQ (N/OFQ) controls several biological functions by selectively activating an opioid like receptor named N/OFQ peptide receptor (NOP). Biased agonism is emerging as an important and therapeutically relevant pharmacological concept in the field of G protein coupled receptors including opioids. To evaluate the relevance of this phenomenon in the NOP receptor, we used a bioluminescence resonance energy transfer technology to measure the interactions of the NOP receptor with either G proteins or β-arrestin 2 in the absence and in presence of increasing concentration of ligands. A large panel of receptor ligands was investigated by comparing their ability to promote or block NOP/G protein and NOP/arrestin interactions. In this study we report a systematic analysis of the functional selectivity of NOP receptor ligands. NOP/G protein interactions (investigated in cell membranes) allowed a precise estimation of both ligand potency and efficacy yielding data highly consistent with the known pharmacological profile of this receptor. The same panel of ligands displayed marked differences in the ability to promote NOP/β-arrestin 2 interactions (evaluated in whole cells). In particular, full agonists displayed a general lower potency and for some ligands an inverted rank order of potency was noted. Most partial agonists behaved as pure competitive antagonists of receptor/arrestin interaction. Antagonists displayed similar values of potency for NOP/Gβ1 or NOP/β-arrestin 2 interaction. Using N/OFQ as reference ligand we computed the bias factors of NOP ligands and a number of agonists with greater efficacy at G protein coupling were identified.


PLOS ONE | 2016

Characterisation of the Novel Mixed Mu-NOP Peptide Ligand Dermorphin-N/OFQ (DeNo)

Mark F. Bird; Maria Camilla Cerlesi; Mark Brown; Davide Malfacini; Vanessa Vezzi; Paola Molinari; Laura Micheli; Lorenzo Di Cesare Mannelli; Carla Ghelardini; Remo Guerrini; Girolamo Calo; David G. Lambert

Introduction Opioid receptors are currently classified as Mu (μ), Delta (δ), Kappa (κ) plus the opioid related nociceptin/orphanin FQ (N/OFQ) peptide receptor (NOP). Despite compelling evidence for interactions and benefits of targeting more than one receptor type in producing analgesia, clinical ligands are Mu agonists. In this study we have designed a Mu-NOP agonist named DeNo. The Mu agonist component is provided by dermorphin, a peptide isolated from the skin of Phyllomedusa frogs and the NOP component by the endogenous agonist N/OFQ. Methods We have assessed receptor binding profile of DeNo and compared with dermorphin and N/OFQ. In a series of functional screens we have assessed the ability to (i) increase Ca2+ in cells coexpressing recombinant receptors and a the chimeric protein Gαqi5, (ii) stimulate the binding of GTPγ[35S], (iii) inhibit cAMP formation, (iv) activate MAPKinase, (v) stimulate receptor-G protein and arrestin interaction using BRET, (vi) electrically stimulated guinea pig ileum (gpI) assay and (vii) ability to produce analgesia via the intrathecal route in rats. Results DeNo bound to Mu (pKi; 9.55) and NOP (pKi; 10.22) and with reasonable selectivity. This translated to increased Ca2+ in Gαqi5 expressing cells (pEC50 Mu 7.17; NOP 9.69), increased binding of GTPγ[35S] (pEC50 Mu 7.70; NOP 9.50) and receptor-G protein interaction in BRET (pEC50 Mu 8.01; NOP 9.02). cAMP formation was inhibited and arrestin was activated (pEC50 Mu 6.36; NOP 8.19). For MAPK DeNo activated p38 and ERK1/2 at Mu but only ERK1/2 at NOP. In the gpI DeNO inhibited electrically-evoked contractions (pEC50 8.63) that was sensitive to both Mu and NOP antagonists. DeNo was antinociceptive in rats. Conclusion Collectively these data validate the strategy used to create a novel bivalent Mu-NOP peptide agonist by combining dermorphin (Mu) and N/OFQ (NOP). This molecule behaves essentially as the parent compounds in vitro. In the antonocicoeptive assays employed in this study DeNo displays only weak antinociceptive properties.


British Journal of Pharmacology | 2014

Pharmacological characterization of tachykinin tetrabranched derivatives

Chiara Ruzza; Anna Rizzi; Davide Malfacini; Maria Camilla Cerlesi; Federica Ferrari; Erika Marzola; Caterina Ambrosio; Cristina Grò; Salvadori Severo; Tommaso Costa; Girolamo Calo; Remo Guerrini

Peptide welding technology (PWT) is a novel chemical strategy that allows the synthesis of multibranched peptides with high yield, purity and reproducibility. Using this technique, we have synthesized and pharmacologically characterized the tetrabranched derivatives of the tachykinins, substance P (SP), neurokinin A (NKA) and B (NKB).


PLOS ONE | 2014

Pharmacological Investigations of N-Substituent Variation in Morphine and Oxymorphone: Opioid Receptor Binding, Signaling and Antinociceptive Activity

Tanila Ben Haddou; Szabolcs Béni; Sándor Hosztafi; Davide Malfacini; Girolamo Calo; Helmut Schmidhammer; Mariana Spetea

Morphine and structurally related derivatives are highly effective analgesics, and the mainstay in the medical management of moderate to severe pain. Pharmacological actions of opioid analgesics are primarily mediated through agonism at the µ opioid peptide (MOP) receptor, a G protein-coupled receptor. Position 17 in morphine has been one of the most manipulated sites on the scaffold and intensive research has focused on replacements of the 17-methyl group with other substituents. Structural variations at the N-17 of the morphinan skeleton led to a diversity of molecules appraised as valuable and potential therapeutics and important research probes. Discovery of therapeutically useful morphine-like drugs has also targeted the C-6 hydroxyl group, with oxymorphone as one of the clinically relevant opioid analgesics, where a carbonyl instead of a hydroxyl group is present at position 6. Herein, we describe the effect of N-substituent variation in morphine and oxymorphone on in vitro and in vivo biological properties and the emerging structure-activity relationships. We show that the presence of a N-phenethyl group in position 17 is highly favorable in terms of improved affinity and selectivity at the MOP receptor, potent agonism and antinociceptive efficacy. The N-phenethyl derivatives of morphine and oxymorphone were very potent in stimulating G protein coupling and intracellular calcium release through the MOP receptor. In vivo, they were highly effective against acute thermal nociception in mice with marked increased antinociceptive potency compared to the lead molecules. It was also demonstrated that a carbonyl group at position 6 is preferable to a hydroxyl function in these N-phenethyl derivatives, enhancing MOP receptor affinity and agonist potency in vitro and in vivo. These results expand the understanding of the impact of different moieties at the morphinan nitrogen on ligand-receptor interaction, molecular mode of action and signaling, and may be instrumental to the development of new opioid therapeutics.


Pharmacology Research & Perspectives | 2017

In vitro pharmacological characterization of a novel unbiased NOP receptor‐selective nonpeptide agonist AT‐403

Federica Ferrari; Davide Malfacini; Blair V. Journigan; Mark F. Bird; Claudio Trapella; Remo Guerrini; David G. Lambert; Girolamo Calo; Nurulain T. Zaveri

Nociceptin/orphanin FQ (N/OFQ) regulates several biological functions via selective activation of the N/OFQ receptor (NOP), a member of the opioid receptor family. We recently identified a new high affinity and highly selective NOP agonist AT‐403. In this study, we characterized the functional profile of AT‐403 and compared it to other known nonpeptide NOP agonists Ro 65‐6570, Ro 2q, SCH‐221510, MCOPPB, AT‐202 and SCH‐486757, using the following assays: GTPγ[35S] stimulated binding, calcium mobilization assay in cells‐expressing human NOP or classical opioid receptors and chimeric G proteins, bioluminescence resonance energy transfer (BRET) based assay for studying NOP receptor interaction with G protein and arrestin, and the electrically stimulated mouse vas deferens bioassay. All compounds behaved as NOP full agonists consistently showing the following rank order of potency MCOPPB > AT‐403 > Ro 65‐6570 = Ro 2q > SCH‐221510 > AT‐202 > SCH‐486757. AT‐403 and MCOPPB displayed the highest NOP selectivity both at human and murine receptors. Interestingly, while all the other nonpeptide NOP agonists displayed bias toward G protein‐mediated signaling in the BRET assay, AT‐403, similar to the natural ligand N/OFQ, behaved as an unbiased agonist, activating G‐protein‐mediated function as well as arrestin recruitment. AT‐403 may be a useful nonpeptide tool compound to study the pharmacology of NOP activation in disease states.

Collaboration


Dive into the Davide Malfacini'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

Mark F. Bird

Leicester Royal Infirmary

View shared research outputs
Researchain Logo
Decentralizing Knowledge