Network


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

Hotspot


Dive into the research topics where Rosaria Luciani is active.

Publication


Featured researches published by Rosaria Luciani.


Scientific Reports | 2013

Discovery of highly potent acid ceramidase inhibitors with in vitro tumor chemosensitizing activity

Natalia Realini; Carlos Solorzano; Chiara Pagliuca; Daniela Pizzirani; Andrea Armirotti; Rosaria Luciani; Maria Paola Costi; Tiziano Bandiera; Daniele Piomelli

The expression of acid ceramidase (AC) – a cysteine amidase that hydrolyses the proapoptotic lipid ceramide – is abnormally high in several human tumors, which is suggestive of a role in chemoresistance. Available AC inhibitors lack, however, the potency and drug-likeness necessary to test this idea. Here we show that the antineoplastic drug carmofur, which is used in the clinic to treat colorectal cancers, is a potent AC inhibitor and that this property is essential to its anti-proliferative effects. Modifications in the chemical scaffold of carmofur yield new AC inhibitors that act synergistically with standard antitumoral drugs to prevent cancer cell proliferation. These findings identify AC as an unexpected target for carmofur, and suggest that this molecule can be used as starting point for the design of novel chemosensitizing agents.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Protein–protein interface-binding peptides inhibit the cancer therapy target human thymidylate synthase

D. Cardinale; Giambattista Guaitoli; Donatella Tondi; Rosaria Luciani; Stefan Henrich; Outi M. H. Salo-Ahen; Stefania Ferrari; Gaetano Marverti; Davide Guerrieri; Alessio Ligabue; Chiara Frassineti; Cecilia Pozzi; Stefano Mangani; D. Fessas; Remo Guerrini; Glauco Ponterini; Rebecca C. Wade; Maria Paola Costi

Human thymidylate synthase is a homodimeric enzyme that plays a key role in DNA synthesis and is a target for several clinically important anticancer drugs that bind to its active site. We have designed peptides to specifically target its dimer interface. Here we show through X-ray diffraction, spectroscopic, kinetic, and calorimetric evidence that the peptides do indeed bind at the interface of the dimeric protein and stabilize its di-inactive form. The “LR” peptide binds at a previously unknown binding site and shows a previously undescribed mechanism for the allosteric inhibition of a homodimeric enzyme. It inhibits the intracellular enzyme in ovarian cancer cells and reduces cellular growth at low micromolar concentrations in both cisplatin-sensitive and -resistant cells without causing protein overexpression. This peptide demonstrates the potential of allosteric inhibition of hTS for overcoming platinum drug resistance in ovarian cancer.


Journal of Medicinal Chemistry | 2011

Virtual screening identification of nonfolate compounds, including a CNS drug, as antiparasitic agents inhibiting pteridine reductase.

Stefania Ferrari; Federica Morandi; Domantas Motiejunas; Erika Nerini; Stefan Henrich; Rosaria Luciani; Alberto Venturelli; Sandra Lazzari; Samuele Calò; Shreedhara Gupta; Véronique Hannaert; Paul A. M. Michels; Rebecca C. Wade; M. Paola Costi

Folate analogue inhibitors of Leishmania major pteridine reductase (PTR1) are potential antiparasitic drug candidates for combined therapy with dihydrofolate reductase (DHFR) inhibitors. To identify new molecules with specificity for PTR1, we carried out a virtual screening of the Available Chemicals Directory (ACD) database to select compounds that could interact with L. major PTR1 but not with human DHFR. Through two rounds of drug discovery, we successfully identified eighteen drug-like molecules with low micromolar affinities and high in vitro specificity profiles. Their efficacy against Leishmania species was studied in cultured cells of the promastigote stage, using the compounds both alone and in combination with 1 (pyrimethamine; 5-(4-chlorophenyl)-6-ethylpyrimidine-2,4-diamine). Six compounds showed efficacy only in combination. In toxicity tests against human fibroblasts, several compounds showed low toxicity. One compound, 5c (riluzole; 6-(trifluoromethoxy)-1,3-benzothiazol-2-ylamine), a known drug approved for CNS pathologies, was active in combination and is suitable for early preclinical evaluation of its potential for label extension as a PTR1 inhibitor and antiparasitic drug candidate.


ACS Chemical Biology | 2015

2-Carboxyquinoxalines Kill Mycobacterium tuberculosis through Noncovalent Inhibition of DprE1

João Neres; Ruben C. Hartkoorn; Laurent R. Chiarelli; Ramakrishna Gadupudi; Maria Rosalia Pasca; Giorgia Mori; Alberto Venturelli; Svetlana Savina; Vadim Makarov; Gaëlle S. Kolly; Elisabetta Molteni; Claudia Binda; Neeraj Dhar; Stefania Ferrari; Priscille Brodin; Vincent Delorme; Valérie Landry; Ana Luisa de Jesus Lopes Ribeiro; Davide Salvatore Francesco Farina; Puneet Saxena; Florence Pojer; Antonio Carta; Rosaria Luciani; Alessio Porta; Giuseppe Zanoni; Edda De Rossi; Maria Paola Costi; Giovanna Riccardi; Stewart T. Cole

Phenotypic screening of a quinoxaline library against replicating Mycobacterium tuberculosis led to the identification of lead compound Ty38c (3-((4-methoxybenzyl)amino)-6-(trifluoromethyl)quinoxaline-2-carboxylic acid). With an MIC99 and MBC of 3.1 μM, Ty38c is bactericidal and active against intracellular bacteria. To investigate its mechanism of action, we isolated mutants resistant to Ty38c and sequenced their genomes. Mutations were found in rv3405c, coding for the transcriptional repressor of the divergently expressed rv3406 gene. Biochemical studies clearly showed that Rv3406 decarboxylates Ty38c into its inactive keto metabolite. The actual target was then identified by isolating Ty38c-resistant mutants of an M. tuberculosis strain lacking rv3406. Here, mutations were found in dprE1, encoding the decaprenylphosphoryl-d-ribose oxidase DprE1, essential for biogenesis of the mycobacterial cell wall. Genetics, biochemical validation, and X-ray crystallography revealed Ty38c to be a noncovalent, noncompetitive DprE1 inhibitor. Structure-activity relationship studies generated a family of DprE1 inhibitors with a range of IC50s and bactericidal activity. Co-crystal structures of DprE1 in complex with eight different quinoxaline analogs provided a high-resolution interaction map of the active site of this extremely vulnerable target in M. tuberculosis.


Journal of Medicinal Chemistry | 2012

Structure-Based Selectivity Optimization of Piperidine–Pteridine Derivatives as Potent Leishmania Pteridine Reductase Inhibitors

Paola Corona; Federica Gibellini; Andrea Cavalli; Puneet Saxena; Antonio Carta; Mario Loriga; Rosaria Luciani; Giuseppe Paglietti; Davide Guerrieri; Erika Nerini; Shreedhara Gupta; Véronique Hannaert; Paul A. M. Michels; Stefania Ferrari; Paola M. Costi

The upregulation of pteridine reductase (PTR1) is a major contributor to antifolate drug resistance in Leishmania spp., as it provides a salvage pathway that bypasses dihydrofolate reductase (DHFR) inhibition. The structure-based optimization of the PTR1 inhibitor methyl-1-[4-(2,4-diaminopteridin-6-ylmethylamino)benzoyl]piperidine-4-carboxylate (1) led to the synthesis of a focused compound library which showed significantly improved selectivity for the parasites folate-dependent enzyme. When used in combination with pyrimethamine, a DHFR inhibitor, a synergistic effect was observed for compound 5b. This work represents a step forward in the identification of effective antileishmania agents.


European Journal of Pharmacology | 2009

Collateral sensitivity to novel thymidylate synthase inhibitors correlates with folate cycle enzymes impairment in cisplatin-resistant human ovarian cancer cells

Gaetano Marverti; Alessio Ligabue; Giuseppe Paglietti; Paola Corona; Sandra Piras; Gabriella Vitale; Davide Guerrieri; Rosaria Luciani; Maria Paola Costi; Chiara Frassineti; Maria Stella Moruzzi

The cytotoxicity of two novel folate cycle inhibitors with quinoxalinic structure, 3-methyl-7-trifluoromethyl-2(R)-[3,4,5-trimethoxyanilino]-quinoxaline (453R) and 3-piperazinilmethyl-2[4(oxymethyl)-phenoxy]quinoxaline (311S), was tested against a panel of both cisplatin(cDDP)-sensitive and -resistant carcinoma cell lines. Interestingly, the cisplatin-resistant human ovarian line, C13 cells, exhibited collateral sensitivity towards the two compounds when compared to its sensitive parental 2008 cells. In this resistant line, which showed elevated expression of the folate cycle enzymes, thymidylate synthase (TS) and dihydrofolate reductase (DHFR), due to cisplatin-resistance phenotype, collateral sensitivity correlated with the greater reduction of enzyme expression. In addition, TS and DHFR expression of the other resistant lines, the human ovarian carcinoma A2780/CP cells and the human breast cancer MDA/CH cells, were decreased in accordance with the similar sensitivity or the low level of cross-resistance to these compounds in comparison to their respective parental lines. Noteworthy, unlike 5-fluorouracil, both drugs reduced the level of TS without inducing ternary complex formation with the co-substrate and the nucleotide analogue. Median effect analysis of the interactive effects of cisplatin with the two quinoxalines mainly showed additive or synergistic cell killing, depending on schedules of drug combinations. In particular, synergistic effects were more often obtained, even on the resistant cells, when cisplatin was added at the beginning of the treatment. These results indicate that, despite the possibility of other mechanisms being involved, inhibition of TS cycle enzymes plays an important role in the pharmacology of these compounds, which might also represent a useful component in drug treatment protocols against cDDP-resistant cells.


Journal of Medicinal Chemistry | 2012

Inhibitor of ovarian cancer cells growth by virtual screening: a new thiazole derivative targeting human thymidylate synthase.

Emanuele Carosati; Anna Tochowicz; Gaetano Marverti; Giambattista Guaitoli; Paolo Benedetti; Stefania Ferrari; Robert M. Stroud; Janet Finer-Moore; Rosaria Luciani; Davide Salvatore Francesco Farina; Gabriele Cruciani; M. Paola Costi

Human thymidylate synthase (hTS) was targeted through a virtual screening approach. The most optimal inhibitor identified, 2-{4-hydroxy-2-[(2-hydroxybenzylidene)hydrazono]-2,5-dihydrothiazol-5-yl}-N-(3-trifluoromethylphenyl)acetamide (5), showed a mixed-type inhibition pattern, with a K(i) of 1.3 μM and activity against ovarian cancer cell lines with the same potency as cisplatin. X-ray studies revealed that it binds the inactive enzyme conformation. This study is the first example of a nonpeptidic inhibitor that binds the inactive hTS and exhibits anticancer activity against ovarian cancer cells.


Journal of Medicinal Chemistry | 2016

Profiling of Flavonol Derivatives for the Development of Antitrypanosomatidic Drugs

Chiara Borsari; Rosaria Luciani; Cecilia Pozzi; Ina Poehner; Stefan Henrich; Matteo Trande; Anabela Cordeiro-da-Silva; Nuno Santarém; Catarina Baptista; Annalisa Tait; Flavio Di Pisa; Lucia Dello Iacono; Giacomo Landi; Sheraz Gul; Markus Wolf; Maria Kuzikov; Bernhard Ellinger; Jeanette Reinshagen; Gesa Witt; Philip Gribbon; Manfred Kohler; Oliver Keminer; Birte Behrens; Luca Costantino; Paloma Tejera Nevado; Eugenia Bifeld; Julia Eick; Joachim Clos; Juan J. Torrado; María Jiménez-Antón

Flavonoids represent a potential source of new antitrypanosomatidic leads. Starting from a library of natural products, we combined target-based screening on pteridine reductase 1 with phenotypic screening on Trypanosoma brucei for hit identification. Flavonols were identified as hits, and a library of 16 derivatives was synthesized. Twelve compounds showed EC50 values against T. brucei below 10 μM. Four X-ray crystal structures and docking studies explained the observed structure-activity relationships. Compound 2 (3,6-dihydroxy-2-(3-hydroxyphenyl)-4H-chromen-4-one) was selected for pharmacokinetic studies. Encapsulation of compound 2 in PLGA nanoparticles or cyclodextrins resulted in lower in vitro toxicity when compared to the free compound. Combination studies with methotrexate revealed that compound 13 (3-hydroxy-6-methoxy-2-(4-methoxyphenyl)-4H-chromen-4-one) has the highest synergistic effect at concentration of 1.3 μM, 11.7-fold dose reduction index and no toxicity toward host cells. Our results provide the basis for further chemical modifications aimed at identifying novel antitrypanosomatidic agents showing higher potency toward PTR1 and increased metabolic stability.


Journal of Medicinal Chemistry | 2014

Optimization of Peptides That Target Human Thymidylate Synthase to Inhibit Ovarian Cancer Cell Growth

Michela Pelà; Puneet Saxena; Rosaria Luciani; Matteo Santucci; Stefania Ferrari; Gaetano Marverti; Chiara Marraccini; Andrea Martello; Silvia Pirondi; Filippo Genovese; Severo Salvadori; Domenico D’Arca; Glauco Ponterini; Maria Paola Costi; Remo Guerrini

Thymidylate synthase (TS) is a target for pemetrexed and the prodrug 5-fluorouracil (5-FU) that inhibit the protein by binding at its active site. Prolonged administration of these drugs causes TS overexpression, leading to drug resistance. The peptide lead, LR (LSCQLYQR), allosterically stabilizes the inactive form of the protein and inhibits ovarian cancer (OC) cell growth with stable TS and decreased dihydrofolate reductase (DHFR) expression. To improve TS inhibition and the anticancer effect, we have developed 35 peptides by modifying the lead. The d-glutamine-modified peptide displayed the best inhibition of cisplatin-sensitive and -resistant OC cell growth, was more active than LR and 5-FU, and showed a TS/DHFR expression pattern similar to LR. Circular dichroism spectroscopy and molecular dynamics studies provided a molecular-level rationale for the differences in structural preferences and the enzyme inhibitory activities. By combining target inhibition studies and the modulation pattern of associated proteins, this work avenues a concept to develop more specific inhibitors of OC cell growth and drug leads.


Journal of Medicinal Chemistry | 2011

Identification of the Binding Modes of N-Phenylphthalimides Inhibiting Bacterial Thymidylate Synthase through X-Ray Crystallography Screening

Stefano Mangani; Laura Cancian; Rosalida Leone; Cecilia Pozzi; Sandra Lazzari; Rosaria Luciani; Stefania Ferrari; Maria Paola Costi

To identify specific bacterial thymidylate synthase (TS) inhibitors, we exploited phenolphthalein (PTH), which inhibits both bacterial and human enzymes. The X-ray crystal structure of Lactobacillus casei TS (LcTS) that binds PTH showed multiple binding modes of the inhibitor, which prevented a classical structure-based drug design approach. To overcome this issue, we synthesized two phthalimidic libraries that were tested against TS enzymes and then we performed X-ray crystallographic screening of the active compounds. Compounds 6A, 8A, and 12A showed 40-fold higher affinity for bacterial TS than human TS. The X-ray crystallographic screening characterized the binding mode of six inhibitors in complexes with LcTS. Of these, 20A, 23A, and 24A showed a common unique binding mode, whereas 8A showed a different, unique binding mode. A comparative analysis of the LcTS X-ray complexes that were obtained with the pathogenic TS enabled the selection of compounds 8A and 23A as specific compounds and starting points to be exploited for the specific inhibition of pathogen enzymes.

Collaboration


Dive into the Rosaria Luciani's collaboration.

Top Co-Authors

Avatar

Maria Paola Costi

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Stefania Ferrari

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gaetano Marverti

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Puneet Saxena

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Alberto Venturelli

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Glauco Ponterini

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Matteo Santucci

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alberto Amaretti

University of Modena and Reggio Emilia

View shared research outputs
Researchain Logo
Decentralizing Knowledge