Network


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

Hotspot


Dive into the research topics where Fabio Arnesano is active.

Publication


Featured researches published by Fabio Arnesano.


Proceedings of the National Academy of Sciences of the United States of America | 2003

A redox switch in CopC: an intriguing copper trafficking protein that binds copper(I) and copper(II) at different sites.

Fabio Arnesano; Lucia Banci; Ivano Bertini; Stefano Mangani; Andrew R. Thompsett

The protein CopC from Pseudomonas syringae has been found capable of binding copper(I) and copper(II) at two different sites, occupied either one at a time or simultaneously. The protein, consisting of 102 amino acids, is known to bind copper(II) in a position that is now found consistent with a coordination arrangement including His-1, Glu-27, Asp-89, and His-91. A full solution structure analysis is reported here for Cu(I)-CopC. The copper(I) site is constituted by His-48 and three of the four Met residues (40, 43, 46, 51), which are clustered in a Met-rich region. Both copper binding sites have been characterized through extended x-ray absorption fine structure studies. They represent novel coordination environments for copper in proteins. The two sites are ≈30 Å far apart and have little affinity for the ion in the other oxidation state. Oxidation of Cu(I)-CopC or reduction of Cu(II)-CopC causes migration of copper from one site to the other. This behavior is observed both in NMR and EXAFS studies and indicates that CopC can exchange copper between two sites activated by a redox switch. CopC resides in the periplasm of Gram-negative bacteria where there is a multicopper oxidase, CopA, which may modulate the redox state of copper. CopC and CopA are coded in the same operon, responsible for copper resistance. These peculiar and novel properties of CopC are discussed with respect to their relevance for copper homeostasis.


Journal of the American Chemical Society | 2011

Probing the interaction of cisplatin with the human copper chaperone Atox1 by solution and in-cell NMR spectroscopy.

Fabio Arnesano; Lucia Banci; Ivano Bertini; Isabella C. Felli; Maurizio Losacco; Giovanni Natile

Among anticancer therapeutics, platinum-based drugs have a prominent role. They carry out their antitumor activity by forming stable adducts with DNA, thus interfering with replication and transcription processes. Cellular uptake of these drugs is tightly connected to copper transport. The major Cu(I) influx transporter Ctr1 has been found to mediate transport of cisplatin and its analogues. Evidence also suggests that ATP7A and ATP7B mediate cisplatin sequestration and efflux from cells, thus influencing drug resistance. The copper-chaperone Atox1, which normally binds Cu(I) via two cysteines and delivers the metal to ATP7A/B, has also been reported to interact with cisplatin in in vitro experiments. In the present investigation we apply a combined approach, using solution and in-cell NMR spectroscopy methods, to probe intracellular drug delivery and interaction of cisplatin with Atox1. The intracellular environment provides itself the suitable conditions for the preservation of the protein in its active form. Initially a {Pt(NH(3))(2)}-Atox1 adduct is formed. At longer reaction time we observed protein dimerization and loss of the ammines. Such a process is reminiscent of the copper-promoted formation of Atox1 dimers which have been proposed to be able to cross the nuclear membrane and act as a transcription factor. We also show that overexpression of Atox1 in E. coli reduces the amount of DNA platination and, consequently, the degree of cell filamentation.


Structure | 2002

Solution structure of CopC: a cupredoxin-like protein involved in copper homeostasis.

Fabio Arnesano; Lucia Banci; Ivano Bertini; Andrew R. Thompsett

The structure of the metal-free form of CopC, a protein involved in copper homeostasis, has been obtained. The fold is a Greek key beta barrel similar to that of functionally unrelated blue copper proteins but with important structural variations. The protein binds one equivalent of copper (II) with relatively high affinity and contains a cluster of conserved residues (His1, Glu27, Asp89, and His91) which could form a water-accessible metal binding site. The structure also reveals a loop containing the M(X)(n)M motif which is present in a number of proteins also involved in copper homeostasis. The present structure represents a link between copper-trafficking proteins and cupredoxins. Within a structural and genomic analysis, the role of CopC in copper trafficking is discussed.


Quarterly Reviews of Biophysics | 2005

NMR structures of paramagnetic metalloproteins

Fabio Arnesano; Lucia Banci; Mario Piccioli

Metalloproteins represent a large share of the proteome and many of them contain paramagnetic metal ions. The knowledge, at atomic resolution, of their structure in solution is important to understand processes in which they are involved, such as electron transfer mechanisms, enzymatic reactions, metal homeostasis and metal trafficking, as well as interactions with their partners. Formerly considered as unfeasible, the first structure in solution by nuclear magnetic resonance (NMR) of a paramagnetic protein was obtained in 1994. Methodological and instrumental advancements pursued over the last decade are such that NMR structure of paramagnetic proteins may be now routinely obtained. We focus here on approaches and problems related to the structure determination of paramagnetic proteins in solution through NMR spectroscopy. After a survey of the background theory, we show how the effects produced by the presence of a paramagnetic metal ion on the NMR parameters, which are in many cases deleterious for the detection of NMR spectra, can be overcome and turned into an additional source of structural restraints. We also briefly address features and perspectives given by the use of 13C-detected protonless NMR spectroscopy for proteins in solution. The structural information obtained through the exploitation of a paramagnetic center are discussed for some Cu2+ -binding proteins and for Ca2+ -binding proteins, where the replacement of a diamagnetic metal ion with suitable paramagnetic metal ions suggests novel approaches to the structural characterization of proteins containing diamagnetic and NMR-silent metal ions.


Angewandte Chemie | 2014

Translocation of Platinum Anticancer Drugs by Human Copper ATPases ATP7A and ATP7B

Francesco Tadini-Buoninsegni; Gianluca Bartolommei; Maria Rosa Moncelli; Giuseppe Inesi; Angela Galliani; Marilù Sinisi; Maurizio Losacco; Giovanni Natile; Fabio Arnesano

Cisplatin, carboplatin, and oxaliplatin are widely used anticancer drugs. Their efficacy is strongly reduced by development of cell resistance. Down-regulation of CTR1 and up-regulation of the Cu-ATPases, ATP7A and ATP7B, have been associated to augmented drug resistance. To gain information on translocation of Pt drugs by human Cu-ATPases, we performed electrical measurements on the COS-1 cell microsomal fraction, enriched with recombinant ATP7A, ATP7B, and selected mutants, and adsorbed on a solid supported membrane. The experimental results indicate that Pt drugs activate Cu-ATPases and undergo ATP-dependent translocation in a fashion similar to that of Cu. We then used NMR spectroscopy and ESI-MS to determine the binding mode of these drugs to the first N-terminal metal-binding domain of ATP7A (Mnk1).


Pure and Applied Chemistry | 2008

Platinum on the road: Interactions of antitumoral cisplatin with proteins

Fabio Arnesano; Giovanni Natile

When the antitumor activity of cisplatin was discovered, no one would have thought of the existence of specific proteins able to transport Pt across the cell membrane or to specifically recognize DNA modified by this drug. However, such proteins do exist and, furthermore, are specific for the Pt substrate considered. It follows that proteins are deeply involved in managing the biological activity of cisplatin. It is expected that, after the first 20 years in which most of the efforts were devoted to understanding its mode of interaction with DNA and consequent structural and functional alterations, the role of proteins will be more deeply scavenged. How cisplatin can survive the attack of the many platinophiles present in the extracellular and intracellular media is the issue addressed in this article. Significantly, differences are observed between cisplatin, carboplatin, and oxaliplatin.


Angewandte Chemie | 2009

Methionine Can Favor DNA Platination by trans‐Coordinated Platinum Antitumor Drugs

Chan Li; Zhanyong Li; Einar Sletten; Fabio Arnesano; Maurizio Losacco; Giovanni Natile; Yangzhong Liu

Cisplatin (cis-DDP) and its cis-coordinated analogues, carboplatin and oxaliplatin, have been successfully used in the treatment of testicular and other solid tumors, but applications are restricted by side effects and intrinsic and acquired resistances. The discovery of trans-coordinated platinum complexes with antitumor activity provides a novel approach for cancer chemotherapy. Among several types of transplatinum complexes, trans-[PtCl2{E-HN=C(OCH3)CH3}2] (trans-EE) raised particular interest because of its higher cytotoxicity than the cis isomer and its activity towards several cis-DDP-resistant tumor cells. Mechanistic studies indicated that trans-EE has different DNA binding modes relative to cis-DDP, although their reaction rates were similar. DNAmodified by trans-EE could not be recognized by high-mobility group (HMG), the protein that interferes with DNA repair of cis-DDP adducts, whereas histone H1 could bind to trans-EE-modified DNA and prevent DNA polymerization and repair. A recent study also indicated that methionine was the preferable binding site of trans-EE in the reaction with cytochrome c, and different binding modes were observed between cisand trans-platinum complexes. Many cellular molecules, including proteins, peptides, and also some small molecules, can play significant roles in the functioning of and resistance to drugs, such as DNA platination, drug transport, and efflux. Sulfur-containing proteins are of special interest because of their high affinity for platinum, their abundance (e.g. albumin), and their involvement in metal-ion transport (e.g. the copper transporter protein CTR1, which contains methionine-rich extracellular motifs and appears to be involved in platinum-drug transport through the cell membrane). Kinetic studies indicated that the S platination of l-methionine (Met) or Nacetyl-l-methionine (AcMet) was kinetically preferred, whereas N7 coordination of guanine was thermodynamically favored. Studies using the model compound [PtCl(dien)] showed that the migration of platinum from S-Met to N7guanine (G–N7) was fairly slow (t1/2= 21–147 h at 310 K depending on DNA sequence), which was obviously slower than the direct DNA platination by [PtCl(dien)]. Although Met could slightly increase the rate of platination of cis-DDP to guanosine monophosphate (GMP), the reaction with synthetic DNA showed that the presence of Met actually inhibited platination on both single strand (ss) and double strand (ds) DNA. Herein we show that the platination rates of both GMP and DNA are substantially enhanced by a Met ligand bound to trans-EE. Moreover the reaction is highly pH-dependent. This enhancement has been observed for all nucleotides used in this work, including monomeric GMP, synthetic ssand dsDNA, and natural DNA. It has been observed that the formation of a Met intermediate is about seven times faster than G–N7 platination (Figure S1 in the Supporting Information). Accordingly, DNA platination is significantly faster via a Met intermediate (Scheme 1). On the basis of activity studies and the formation of this type of adducts also in the cellular system, it is suggested that the mechanism of trans-EE could differ substantially from that of conventional cisplatinum compounds.


PLOS ONE | 2009

Copper-Triggered Aggregation of Ubiquitin

Fabio Arnesano; Simone Scintilla; Vincenza Calò; Elena Bonfrate; Chiara Ingrosso; Maurizio Losacco; Teresa Pellegrino; Enrico Rizzarelli; Giovanni Natile

Neurodegenerative disorders share common features comprising aggregation of misfolded proteins, failure of the ubiquitin-proteasome system, and increased levels of metal ions in the brain. Protein aggregates within affected cells often contain ubiquitin, however no report has focused on the aggregation propensity of this protein. Recently it was shown that copper, differently from zinc, nickel, aluminum, or cadmium, compromises ubiquitin stability and binds to the N-terminus with 0.1 micromolar affinity. This paper addresses the role of copper upon ubiquitin aggregation. In water, incubation with Cu(II) leads to formation of spherical particles that can progress from dimers to larger conglomerates. These spherical oligomers are SDS-resistant and are destroyed upon Cu(II) chelation or reduction to Cu(I). In water/trifluoroethanol (80∶20, v/v), a mimic of the local decrease in dielectric constant experienced in proximity to a membrane surface, ubiquitin incubation with Cu(II) causes time-dependent changes in circular dichroism and Fourier-transform infrared spectra, indicative of increasing β-sheet content. Analysis by atomic force and transmission electron microscopy reveals, in the given order, formation of spherical particles consistent with the size of early oligomers detected by gel electrophoresis, clustering of these particles in straight and curved chains, formation of ring structures, growth of trigonal branches from the rings, coalescence of the trigonal branched structures in a network. Notably, none of these ubiquitin aggregates was positive to tests for amyloid and Cu(II) chelation or reduction produced aggregate disassembly. The early formed Cu(II)-stabilized spherical oligomers, when reconstituted in 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) liposomes and in POPC planar bilayers, form annular and pore-like structures, respectively, which are common to several neurodegenerative disorders including Parkinsons, Alzheimers, amyotrophic lateral sclerosis, and prion diseases, and have been proposed to be the primary toxic species. Susceptibility to aggregation of ubiquitin, as it emerges from the present study, may represent a potential risk factor for disease onset or progression while cells attempt to tag and process toxic substrates.


European Journal of Pharmaceutics and Biopharmaceutics | 2015

Intranasal delivery of dopamine to the striatum using glycol chitosan/sulfobutylether-β-cyclodextrin based nanoparticles.

Sante Di Gioia; Adriana Trapani; Delia Mandracchia; Elvira De Giglio; S. Cometa; Vincenzo Mangini; Fabio Arnesano; Giuliana Belgiovine; Stefano Castellani; Lorenzo Pace; Michele Angelo Lavecchia; Giuseppe Trapani; Massimo Conese; Giovanni Puglisi; Tommaso Cassano

The aim of this study was to evaluate chitosan (CS)-, glycol chitosan (GCS)- and corresponding thiomer-based nanoparticles (NPs) for delivering dopamine (DA) to the brain by nasal route. Thus, the polyanions tripolyphosphate and sulfobutylether-β-cyclodextrin (SBE-β-CD), respectively, were used as polycation crosslinking agents and SBE-β-CD also in order to enhance the DA stability. The most interesting formulation, containing GCS and SBE-β-CD, was denoted as DA GCS/DA-CD NPs. NMR spectroscopy demonstrated an inclusion complex formation between SBE-β-CD and DA. X-ray photoelectron spectroscopy analysis revealed the presence of DA on the external surface of NPs. DA GCS/DA-CD NPs showed cytotoxic effect toward Olfactory Ensheathing Cells only at higher dosage. Acute administration of DA GCS/DA-CD NPs into the right nostril of rats did not modify the levels of the neurotransmitter in both right and left striatum. Conversely, repeated intranasal administration of DA GCS/DA-CD NPs into the right nostril significantly increased DA in the ipsilateral striatum. Fluorescent microscopy of olfactory bulb after acute administration of DA fluorescent-labeled GCS/DA-CD NPs into the right nostril showed the presence of NPs only in the right olfactory bulb and no morphological tissue damage occurred. Thus, these GCS based NPs could be potentially used as carriers for nose-to-brain DA delivery for the Parkinsons disease treatment.


Journal of Medicinal Chemistry | 2009

Mechanistic insight into the inhibition of matrix metalloproteinases by platinum substrates.

Fabio Arnesano; Angela Boccarelli; Daniele Cornacchia; Fiorentin Nushi; Rossella Sasanelli; Mauro Coluccia; Giovanni Natile

Platinum compounds are among the most used DNA-damaging anticancer drugs, however they can also be tailored to target biological substrates different from DNA, for instance enzymes involved in cancer progression. We recently reported that some platinum complexes with three labile ligands inhibit matrix metalloproteinase activity in a selective way. We have now extended the investigation to a series of platinum complexes having three chlorido or one chlorido and a dimethylmalonato leaving ligands. All compounds are strong inhibitors of MMP-3 by a noncompetitive mechanism, while platinum drugs in clinical use are not. Structural investigations reveal that the platinum substrate only loses two labile ligands, which are replaced by an imidazole nitrogen of His224 and a hydroxyl group, while it retains one chlorido ligand. A chlorido and a hydroxyl group are also present in the zinc complex inhibitor of carboxypeptidase A, whose active site has strong analogies with that of MMP-3.

Collaboration


Dive into the Fabio Arnesano's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lucia Banci

University of Florence

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yangzhong Liu

University of Science and Technology of China

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paolo Carloni

Forschungszentrum Jülich

View shared research outputs
Researchain Logo
Decentralizing Knowledge