Network


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

Hotspot


Dive into the research topics where Angela Arciello is active.

Publication


Featured researches published by Angela Arciello.


Cancer Research | 2004

A Fully Human Antitumor ImmunoRNase Selective for ErbB-2-Positive Carcinomas

Claudia De Lorenzo; Angela Arciello; Rosanna Cozzolino; Donald B. Palmer; Paolo Laccetti; Renata Piccoli; Giuseppe D'Alessio

We report the preparation and characterization of a novel, fully human antitumor immunoRNase (IR). The IR, a human RNase and fusion protein made up of a human single chain variable fragment (scFv), is directed to the ErbB-2 receptor and overexpressed in many carcinomas. The anti-ErbB-2 IR, named hERB-hRNase, retains the enzymatic activity of the wild-type enzyme (human pancreatic RNase) and specifically binds to ErbB-2-positive cells with the high affinity (Kd = 4.5 nm) of the parental scFv. hERB-hRNase behaves as an immunoprotoxin and on internalization by target cells becomes selectively cytotoxic in a dose-dependent manner at nanomolar concentrations. Administered in five doses of 1.5 mg/kg to mice bearing an ErbB-2-positive tumor, hERB-hRNase induced a dramatic reduction in tumor volume. hERB-hRNase is the first fully human antitumor IR produced thus far, with a high potential as a poorly immunogenic human drug devoid of nonspecific toxicity, directed against ErbB-2-positive malignancies.


Journal of Molecular Biology | 2011

Effects of the Known Pathogenic Mutations on the Aggregation Pathway of the Amyloidogenic Peptide of Apolipoprotein A-I

Sara Raimondi; Fulvio Guglielmi; Sofia Giorgetti; Sonia Di Gaetano; Angela Arciello; Daria Maria Monti; Annalisa Relini; Daniela Nichino; Silvia Maria Doglia; Antonino Natalello; Piero Pucci; Palma Mangione; Laura Obici; Giampaolo Merlini; Monica Stoppini; Paul Robustelli; Gian Gaetano Tartaglia; Michele Vendruscolo; Christopher M. Dobson; Renata Piccoli; Vittorio Bellotti

The 93-residue N-terminal fragment of apolipoprotein A-I (ApoA-I) is the major constituent of fibrils isolated from patients affected by the amyloidosis caused by ApoA-I mutations. We have prepared eight polypeptides corresponding to all the currently known amyloidogenic variants of the N-terminal region of ApoA-I, other than a truncation mutation, and investigated their aggregation kinetics and the associated structural modifications. All the variants adopted a monomeric highly disordered structure in solution at neutral pH, whereas acidification of the solution induced an unstable α-helical conformation and the subsequent aggregation into the cross-β structure aggregate. Two mutations (Δ70-72 and L90P) almost abrogated the lag phase of the aggregation process, three mutations (Δ60-71, L75P, and W50R) significantly accelerated the aggregation rate by 2- to 3-fold, while the remaining three variants (L64P, L60R, and G26R) were not significantly different from the wild type. Therefore, an increase in aggregation propensity cannot explain per se the mechanism of the disease for all the variants. Prediction of the protection factors for hydrogen exchange in the native state of full-length protein reveals, in almost all the variants, an expansion of the conformational fluctuations that could favour the proteolytic cleavage and the release of the amyloidogenic peptide. Such an event seems to be a necessary prerequisite for ApoA-I fibrillogenesis in vivo, but the observed increased aggregation propensity of certain variants can have a strong influence on the severity of the disease, such as an earlier onset and a faster progression.


Journal of Biotechnology | 2015

Biocompatibility, uptake and endocytosis pathways of polystyrene nanoparticles in primary human renal epithelial cells.

Daria Maria Monti; Daniela Guarnieri; Giuliana Napolitano; Renata Piccoli; Paolo A. Netti; Sabato Fusco; Angela Arciello

Recent years have witnessed an unprecedented growth in the number of applications—such as drug delivery, nutraceuticals and production of improved biocompatible materials—in the areas of nanoscience and nanotechnology. Engineered nanoparticles (NPs) are an important tool for the development of quite a few of these applications. Despite intense research activity, mechanisms regulating the uptake of NPs into cells are not completely defined, being the phenomenon dramatically influenced by physico-chemical properties of NPs and cell-specific differences. Since the cellular uptake of NPs is a prerequisite for their use in nanomedicine, the definition of their internalization pathway is crucial. For this reason, we used 44 nm polystyrene NPs as a model to analyze the uptake and endocytosis pathways in primary human renal cortical epithelial (HRCE) cells, which play a key role in the clearance of drugs. NPs were found not to affect the viability and cell cycle progression of HRCE cells. Distinct internalization pathways were analyzed by the use of drugs known to inhibit specific endocytosis routes. Analyses, performed by confocal microscopy in combination with quantitative spectrofluorimetric assays, indicated that NPs enter HRCE cells through multiple mechanisms, either energy-dependent (endocytosis) or energy-independent.


Biological Chemistry | 2013

Human carbonic anhydrase VII protects cells from oxidative damage.

Rita Del Giudice; Daria Maria Monti; Emanuela Truppo; Angela Arciello; Claudiu T. Supuran; Giuseppina De Simone; Simona Maria Monti

Abstract Human carbonic anhydrase (hCA) VII is a cytosolic enzyme with high carbon dioxide hydration activity. Recently, S-glutathionylation of two cysteine residues from the enzyme was revealed, suggesting a new role as oxygen radical scavenger. We analyzed the effect of native and tetramutated hCA VII (all cysteines mutated into serines) in a eukaryotic system by stressing cells with an oxidant agent. Results clearly show that native hCA VII can protect cells from oxidative damage by preventing the apoptosis cascade and that cysteines play a leading role in this process. Our findings definitively confirm hCA VII protective role toward oxidative insult.


Journal of Cellular and Molecular Medicine | 2011

Insights into the fate of the N-terminal amyloidogenic polypeptide of ApoA-I in cultured target cells

Angela Arciello; Nadia De Marco; Rita Del Giudice; Fulvio Guglielmi; Piero Pucci; Annalisa Relini; Daria Maria Monti; Renata Piccoli

Apolipoprotein A‐I (ApoA‐I) is an extracellular lipid acceptor, whose role in cholesterol efflux and high‐density lipoprotein formation is mediated by ATP‐binding cassette transporter A1 (ABCA1). Nevertheless, some ApoA‐I variants are associated to systemic forms of amyloidosis, characterized by extracellular fibril deposition in peripheral organs. Heart amyloid fibrils were found to be mainly constituted by the 93‐residue N‐terminal fragment of ApoA‐I, named [1–93]ApoA‐I. In this paper, rat cardiomyoblasts were used as target cells to analyse binding, internalization and intracellular fate of the fibrillogenic polypeptide in comparison to full‐length ApoA‐I. We provide evidence that the polypeptide: (i) binds to specific sites on cell membrane (Kd= 5.90 ± 0.70 × 10−7 M), where it partially co‐localizes with ABCA1, as also described for ApoA‐I; (ii) is internalized mostly by chlatrin‐mediated endocytosis and lipid rafts, whereas ApoA‐I is internalized preferentially by chlatrin‐coated pits and macropinocytosis and (iii) is rapidly degraded by proteasome and lysosomes, whereas ApoA‐I partially co‐localizes with recycling endosomes. Vice versa, amyloid fibrils, obtained by in vitro aggregation of [1–93]ApoA‐I, were found to be unable to enter the cells. We propose that internalization and intracellular degradation of [1–93]ApoA‐I may divert the polypeptide from amyloid fibril formation and contribute to the slow progression and late onset that characterize this pathology.


Biomaterials | 2009

Enzymatically active fibrils generated by the self-assembly of the ApoA-I fibrillogenic domain functionalized with a catalytic moiety

Fulvio Guglielmi; Daria Maria Monti; Angela Arciello; Silvia Torrassa; Flora Cozzolino; Piero Pucci; Annalisa Relini; Renata Piccoli

Enzymatically active fibrils were produced by self-assembly of a bifunctional chimeric protein, made up of a fibrillogenic and a catalytic moiety. For this purpose, the fibrillogenic domain of Apolipoprotein A-I (ApoA-I), a 93-residue polypeptide named [1-93]ApoA-I, was functionalized with the enzyme glutathione S-transferase (GST). The fusion protein GST-[1-93]ApoA-I was expressed, isolated to homogeneity and characterized. In the soluble form, GST-[1-93]ApoA-I was found to be fully active as a GST enzyme, and to have high propensity to self-aggregate. Upon incubation for 3 weeks at pH 6.4, insoluble aggregates were generated. Analyzed by AFM, they were found to contain fibrillar structures often organized into large fiber networks. Fibrils were loaded on the membrane of a microfiltration unit and tested for enzymatic activity by filtering the substrate through the fibrillar network. Fibrils were shown to be catalytically active, stable over time and reusable, as no loss of activity was detected when fibrils were repeatedly tested. Our findings suggest that catalytically active fibrils may be of interest for biocatalytic applications in nanobiotechnology.


Biochimica et Biophysica Acta | 2016

Protein conformational perturbations in hereditary amyloidosis: Differential impact of single point mutations in ApoAI amyloidogenic variants

Rita Del Giudice; Angela Arciello; Francesco Itri; Antonello Merlino; Maria Gaia Monti; Martina Buonanno; Amanda Penco; Diana Canetti; Ganna Petruk; Simona Maria Monti; Annalisa Relini; Piero Pucci; Renata Piccoli; Daria Maria Monti

Amyloidoses are devastating diseases characterized by accumulation of misfolded proteins which aggregate in fibrils. Specific gene mutations in Apolipoprotein A I (ApoAI) are associated with systemic amyloidoses. Little is known on the effect of mutations on ApoAI structure and amyloid properties. Here we performed a physico-chemical characterization of L75P- and L174S-amyloidogenic ApoAI (AApoAI) variants to shed light on the effects of two single point mutations on protein stability, proteolytic susceptibility and aggregation propensity. Both variants are destabilized in their N-terminal region and generate fibrils with different morphological features. L75P-AApoAI is significantly altered in its conformation and compactness, whereas a more flexible and pronounced aggregation-competent state is associated to L174S-AApoAI. These observations point out how single point mutations in ApoAI gene evocate differences in the physico-chemical and conformational behavior of the corresponding protein variants, with the common feature of diverting ApoAI from its natural role towards a pathogenic pathway.


Protein Science | 2008

Structural features for the mechanism of antitumor action of a dimeric human pancreatic ribonuclease variant.

Antonello Merlino; Giovanna Avella; Sonia Di Gaetano; Angela Arciello; Renata Piccoli; Lelio Mazzarella; Filomena Sica

A specialized class of RNases shows a high cytotoxicity toward tumor cell lines, which is critically dependent on their ability to reach the cytosol and to evade the action of the ribonuclease inhibitor (RI). The cytotoxicity and antitumor activity of bovine seminal ribonuclease (BSRNase), which exists in the native state as an equilibrium mixture of a swapped and an unswapped dimer, are peculiar properties of the swapped form. A dimeric variant (HHP2‐RNase) of human pancreatic RNase, in which the enzyme has been engineered to reproduce the sequence of BSRNase helix‐II (Gln28→Leu, Arg31→Cys, Arg32→Cys, and Asn34→Lys) and to eliminate a negative charge on the surface (Glu111→Gly), is also extremely cytotoxic. Surprisingly, this activity is associated also to the unswapped form of the protein. The crystal structure reveals that on this molecule the hinge regions, which are highly disordered in the unswapped form of BSRNase, adopt a very well‐defined conformation in both subunits. The results suggest that the two hinge peptides and the two Leu28 side chains may provide an anchorage to a transient noncovalent dimer, which maintains Cys31 and Cys32 of the two subunits in proximity, thus stabilizing a quaternary structure, similar to that found for the noncovalent swapped dimer of BSRNase, that allows the molecule to escape RI and/or to enhance the formation of the interchain disulfides.


Cellular and Molecular Life Sciences | 2016

Femtosecond UV-laser pulses to unveil protein–protein interactions in living cells

Francesco Itri; Daria Maria Monti; Bartolomeo Della Ventura; Marco Chino; Felice Gesuele; A. Lombardi; R. Velotta; C. Altucci; Leila Birolo; Renata Piccoli; Angela Arciello

A hallmark to decipher bioprocesses is to characterize protein–protein interactions in living cells. To do this, the development of innovative methodologies, which do not alter proteins and their natural environment, is particularly needed. Here, we report a method (LUCK, Laser UV Cross-linKing) to in vivo cross-link proteins by UV-laser irradiation of living cells. Upon irradiation of HeLa cells under controlled conditions, cross-linked products of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were detected, whose yield was found to be a linear function of the total irradiation energy. We demonstrated that stable dimers of GAPDH were formed through intersubunit cross-linking, as also observed when the pure protein was irradiated by UV-laser in vitro. We proposed a defined patch of aromatic residues located at the enzyme subunit interface as the cross-linking sites involved in dimer formation. Hence, by this technique, UV-laser is able to photofix protein surfaces that come in direct contact. Due to the ultra-short time scale of UV-laser-induced cross-linking, this technique could be extended to weld even transient protein interactions in their native context.


Biotechnology Letters | 2011

Expression of human apolipoprotein A-I in Nicotiana tabacum.

Pasquale Chiaiese; Maria Minutolo; Angela Arciello; Fulvio Guglielmini; Renata Piccoli; Edgardo Filippone

Several transgenic tobacco lines expressing human apolipoprotein A-I (ApoA-I) were obtained. Western blot analyses indicated the expression of the recombinant protein in plant organs at various stages of development, including senescent leaves. A cell line expressing human ApoA-I was established from a T1 transgenic plant. Recombinant ApoA-I was isolated either from extracts of transgenic leaves and from the culture medium of transgenic cells using an antibody-based one-step procedure.

Collaboration


Dive into the Angela Arciello's collaboration.

Top Co-Authors

Avatar

Renata Piccoli

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Daria Maria Monti

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Rosa Gaglione

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fulvio Guglielmi

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Piero Pucci

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Eugenio Notomista

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Andrea Bosso

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Elio Pizzo

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Francesco Itri

University of Naples Federico II

View shared research outputs
Researchain Logo
Decentralizing Knowledge