Network


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

Hotspot


Dive into the research topics where Maria Paola Costi is active.

Publication


Featured researches published by Maria Paola Costi.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Discovery of potent pteridine reductase inhibitors to guide antiparasite drug development

Antonio Cavazzuti; Giuseppe Paglietti; William N. Hunter; Francisco Gamarro; Sandra Piras; Mario Loriga; Sergio Allecca; Paola Corona; Karen McLuskey; Lindsay B. Tulloch; Federica Gibellini; Stefania Ferrari; Maria Paola Costi

Pteridine reductase (PTR1) is essential for salvage of pterins by parasitic trypanosomatids and is a target for the development of improved therapies. To identify inhibitors of Leishmania major and Trypanosoma cruzi PTR1, we combined a rapid-screening strategy using a folate-based library with structure-based design. Assays were carried out against folate-dependent enzymes including PTR1, dihydrofolate reductase (DHFR), and thymidylate synthase. Affinity profiling determined selectivity and specificity of a series of quinoxaline and 2,4-diaminopteridine derivatives, and nine compounds showed greater activity against parasite enzymes compared with human enzymes. Compound 6a displayed a Ki of 100 nM toward LmPTR1, and the crystal structure of the LmPTR1:NADPH:6a ternary complex revealed a substrate-like binding mode distinct from that previously observed for similar compounds. A second round of design, synthesis, and assay produced a compound (6b) with a significantly improved Ki (37 nM) against LmPTR1, and the structure of this complex was also determined. Biological evaluation of selected inhibitors was performed against the extracellular forms of T. cruzi and L. major, both wild-type and overexpressing PTR1 lines, as a model for PTR1-driven antifolate drug resistance and the intracellular form of T. cruzi. An additive profile was observed when PTR1 inhibitors were used in combination with known DHFR inhibitors, and a reduction in toxicity of treatment was observed with respect to administration of a DHFR inhibitor alone. The successful combination of antifolates targeting two enzymes indicates high potential for such an approach in the development of previously undescribed antiparasitic drugs.


Current Medicinal Chemistry | 2005

Thymidylate Synthase Structure, Function and Implication in Drug Discovery

Maria Paola Costi; Stefania Ferrari; Alberto Venturelli; Samuele Calò; Donatella Tondi; Daniela Barlocco

Recent methodologies applied to the drug discovery process, such as genomics and proteomics, have greatly implemented our basic understanding of drug action and are giving more input to medicinal chemists, in finding genuinely new targets and opportunities for the development of drugs with original mechanisms of action. In this paper, an example of the successful application of some new techniques to the target enzymes with the Thymidylate Synthase (TS) function is given. The improved knowledge of the complex mechanism of the biological pathways in which thymidylate synthase is involved represents a unique chance to find new mechanism-based inhibitors, aimed to treat not only cancerous diseases, but also infectious pathologies. Thymidylate synthase (TS or ThyA) has long been considered as one of the best-known drug targets in the anti-cancer area, after which old and new drugs, such as 5-fluoro uracil and the anti-folate ZD1694, have been introduced into chemotherapy to treat solid tumours. Only a few attempts have been made to find non-classical anti-folate inhibitors that are dissimilar to the folate co-factor, with the aim of finding unshared protein target domains on the enzyme structure, in order to specifically inhibit TS enzymes from pathogens. Only recently from omic studies, a new Thymidylate Synthase Complementing Protein (TSCP or ThyX) has been identified in a number of pathogens, showing a different structure with respect to human TS, thus opening new avenues to specific inhibitions. A depiction of the most recent progress in the study of Thymidylate Synthase enzymes is presented in the following sections.


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.


Current Drug Targets | 2001

Update on Antifolate Drugs Targets

Maria Paola Costi; Stefania Ferrari

Antifolate drugs are molecules directed to interfere with the folate metabolic pathway at some level. They can be recognized among the first rationally designed compounds applying the principle of structural analogy with the substrate developing the antimetabolite strategy. This strategy has taken advantage of the basic different features of the microbial and human folate metabolism and therefore allows targeting the pathway at different steps generating a specificity tools for Medicinal Chemists. Two main problems are giving renewed importance to such targets and therefore improving the efforts to discover new targets in the folate metabolism area. The first one is the increasing resistance to the present drugs due to different mechanisms such as the enzyme modification and the increased production of enzymes with not well recognized importance. The second one is the development of techniques directed to highlight the interference at genetic level of molecular probes as antifolate drug to develop new enzymes previously unknown. This approach is defined as genetic approach to drug discovery, from gene to drugs. The present article describes the importance in drug design and discovery of some antifolate targets among the best known at the present status of research such as thymidylate synthase (TS), dhydrofolate reductases, (DHFR) serine hydroxymethyltransferase (SHMT), folyilpolyglutamyl synthetase (FPGS), gamma-glutamyl hydrolase (gamma-GH), glycinamide-ribonucleotide transformylase (GARTfase), amino-imidazole-carboxamide-ribonucleotide transformylase (AICARTfase) and Folate transporters. Discovery, known functions, structure/function studies and inhibition will be described.


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 | 2015

The Hippo Pathway and YAP/TAZ–TEAD Protein–Protein Interaction as Targets for Regenerative Medicine and Cancer Treatment

Matteo Santucci; Tatiana Vignudelli; Stefania Ferrari; Marco Mor; Laura Scalvini; Maria Laura Bolognesi; Elisa Uliassi; Maria Paola Costi

The Hippo pathway is an important organ size control signaling network and the major regulatory mechanism of cell-contact inhibition. Yes associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are its targets and terminal effectors: inhibition of the pathway promotes YAP/TAZ translocation to the nucleus, where they interact with transcriptional enhancer associate domain (TEAD) transcription factors and coactivate the expression of target genes, promoting cell proliferation. Defects in the pathway can result in overgrowth phenotypes due to deregulation of stem-cell proliferation and apoptosis; members of the pathway are directly involved in cancer development. The pharmacological regulation of the pathway might be useful in cancer prevention, treatment, and regenerative medicine applications; currently, a few compounds can selectively modulate the pathway. In this review, we present an overview of the Hippo pathway, the sequence and structural analysis of YAP/TAZ, the known pharmacological modulators of the pathway, especially those targeting YAP/TAZ-TEAD interaction.


Current Medicinal Chemistry | 2010

Homodimeric enzymes as drug targets.

D. Cardinale; Outi M. H. Salo-Ahen; Stefania Ferrari; Glauco Ponterini; Gabriele Cruciani; Emanuele Carosati; Anna Tochowicz; Stefano Mangani; Rebecca C. Wade; Maria Paola Costi

Many enzymes and proteins are regulated by their quaternary structure and/or by their association in homo- and/or hetero-oligomer complexes. Thus, these protein-protein interactions can be good targets for blocking or modulating protein function therapeutically. The large number of oligomeric structures in the Protein Data Bank (http://www.rcsb.org/) reflects growing interest in proteins that function as multimeric complexes. In this review, we consider the particular case of homodimeric enzymes as drug targets. There is intense interest in drugs that inhibit dimerization of a functionally obligate homodimeric enzyme. Because amino acid conservation within enzyme interfaces is often low compared to conservation in active sites, it may be easier to achieve drugs that target protein interfaces selectively and specifically. Two main types of dimerization inhibitors have been developed: peptides or peptidomimetics based on sequences involved in protein-protein interactions, and small molecules that act at hot spots in protein-protein interfaces. Examples include inhibitors of HIV protease and HIV integrase. Studying the mechanisms of action and locating the binding sites of such inhibitors requires different techniques for different proteins. For some enzymes, ligand binding is only detectable in vivo or after unfolding of the complexes. Here, we review the structural features of dimeric enzymes and give examples of inhibition through interference in dimer stability. Several techniques for studying these complex phenomena will be presented.


Chemistry & Biology | 2001

Structure-based design and in-parallel synthesis of inhibitors of AmpC β-lactamase

Donatella Tondi; Rachel A. Powers; Emilia Caselli; María-Cristina Negri; Jesús Blázquez; Maria Paola Costi; Brian K. Shoichet

Abstract Background: Group I β-lactamases are a major cause of antibiotic resistance to β-lactams such as penicillins and cephalosporins. These enzymes are only modestly affected by classic β-lactam-based inhibitors, such as clavulanic acid. Conversely, small arylboronic acids inhibit these enzymes at sub-micromolar concentrations. Structural studies suggest these inhibitors bind to a well-defined cleft in the group I β-lactamase AmpC; this cleft binds the ubiquitous R1 side chain of β-lactams. Intriguingly, much of this cleft is left unoccupied by the small arylboronic acids. Results: To investigate if larger boronic acids might take advantage of this cleft, structure-guided in-parallel synthesis was used to explore new inhibitors of AmpC. Twenty-eight derivatives of the lead compound, 3-aminophenylboronic acid, led to an inhibitor with 80-fold better binding ( 2 ; K i 83 nM). Molecular docking suggested orientations for this compound in the R1 cleft. Based on the docking results, 12 derivatives of 2 were synthesized, leading to inhibitors with K i values of 60 nM and with improved solubility. Several of these inhibitors reversed the resistance of nosocomial Gram-positive bacteria, though they showed little activity against Gram-negative bacteria. The X-ray crystal structure of compound 2 in complex with AmpC was subsequently determined to 2.1 A resolution. The placement of the proximal two-thirds of the inhibitor in the experimental structure corresponds with the docked structure, but a bond rotation leads to a distinctly different placement of the distal part of the inhibitor. In the experimental structure, the inhibitor interacts with conserved residues in the R1 cleft whose role in recognition has not been previously explored. Conclusions: Combining structure-based design with in-parallel synthesis allowed for the rapid exploration of inhibitor functionality in the R1 cleft of AmpC. The resulting inhibitors differ considerably from β-lactams but nevertheless inhibit the enzyme well. The crystal structure of 2 ( K i 83 nM) in complex with AmpC may guide exploration of a highly conserved, largely unexplored cleft, providing a template for further design against AmpC β-lactamase.


Bioorganic & Medicinal Chemistry Letters | 2010

Structural study of phenyl boronic acid derivatives as AmpC β-lactamase inhibitors

Donatella Tondi; Samuele Calò; Brian K. Shoichet; Maria Paola Costi

A small set of boronic acids acting as low nanomolar inhibitors of AmpC beta-lactamase were designed and synthesized in the effort to improve affinity, pharmacokinetic properties, and to provide a valid lead compound. X-ray crystallography revealed the binary complex of the best inhibitor bound to the enzyme, highlighting possibilities for its further rational derivatization and chemical optimization.

Collaboration


Dive into the Maria Paola Costi's collaboration.

Top Co-Authors

Avatar

Stefania Ferrari

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

Donatella Tondi

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

Rosaria Luciani

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Gaetano Marverti

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

Filippo Genovese

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge