Network


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

Hotspot


Dive into the research topics where Barbara Lelli is active.

Publication


Featured researches published by Barbara Lelli.


Antimicrobial Agents and Chemotherapy | 2005

Antimicrobial Activity of Novel Dendrimeric Peptides Obtained by Phage Display Selection and Rational Modification

Alessandro Pini; Andrea Giuliani; Chiara Falciani; Ylenia Runci; Claudia Ricci; Barbara Lelli; Monica Malossi; Paolo Neri; Gian Maria Rossolini; Luisa Bracci

ABSTRACT A large 10-mer phage peptide library was panned against whole Escherichia coli cells, and an antimicrobial peptide (QEKIRVRLSA) was selected. The peptide was synthesized in monomeric and dendrimeric tetrabranched form (multiple antigen peptide [MAP]), which generally allows a dramatic increase of peptide stability to peptidases and proteases. The antibacterial activity of the dendrimeric peptide against E. coli was much higher than that of the monomeric form. Modification of the original sequence, by residue substitution or sequence shortening, produced three different MAPs, M4 (QAKIRVRLSA), M5 (KIRVRLSA), and M6 (QKKIRVRLSA) with enhanced stability to natural degradation and antimicrobial activity against a large panel of gram-negative bacteria. The MICs of the most potent peptide, M6, were as low as 4 to 8 μg/ml against recent clinical isolates of multidrug-resistant Pseudomonas aeruginosa and members of the Enterobacteriaceae. The same dendrimeric peptides showed high stability to blood proteases, low hemolytic activity, and low cytotoxic effects on eukaryotic cells, making them promising candidates for the development of new antibacterial drugs.


Chemical Biology & Drug Design | 2007

Molecular Basis of Branched Peptides Resistance to Enzyme Proteolysis

Chiara Falciani; Luisa Lozzi; Alessandro Pini; Federico Corti; Monica Fabbrini; Andrea Bernini; Barbara Lelli; Neri Niccolai; Luisa Bracci

We found that synthetic peptides in the form of dendrimers become resistant to proteolysis. To determine the molecular basis of this resistance, different bioactive peptides were synthesized in monomeric, two‐branched and tetra‐branched form and incubated with human plasma and serum. Proteolytic resistance of branched multimeric sequences was compared to that of the same peptides synthesized as multimeric linear molecules. Unmodified peptides and cleaved sequences were detected by high pressure liquid chromatography and mass spectrometry. An increase in peptide copies did not increase peptide resistance in linear multimeric sequences, whereas multimericity progressively enhanced proteolytic stability of branched multimeric peptides. A structure‐based hypothesis of branched peptide resistance to proteolysis by metallopeptidases is presented.


Molecular Cancer Therapeutics | 2007

Synthesis and biological activity of stable branched neurotensin peptides for tumor targeting

Chiara Falciani; Monica Fabbrini; Alessandro Pini; Luisa Lozzi; Barbara Lelli; Silvia Pileri; Jlenia Brunetti; Stefano Bindi; Silvia Scali; Luisa Bracci

Receptors for endogenous regulatory peptides, like the neuropeptide neurotensin, are overexpressed in several human cancers and can be targets for peptide-mediated tumor-selective therapy. Peptides, however, have the main drawback of an extremely short half-life in vivo. We showed that neurotensin and other endogenous peptides, when synthesized as dendrimers, retain biological activity and become resistant to proteolysis. Here, we synthesized the neurotensin functional fragment NT(8-13) in a tetrabranched form linked to different units for tumor therapy or diagnosis. Fluorescent molecules were used to monitor receptor binding and internalization in HT29 human adenocarcinoma cells and receptor binding in HT29 tumor xenografts in nude mice. Linking of chemotherapic molecules like chlorin e6 and methotrexate to dendrimers resulted in a dramatic increase in drug selectivity, uptake of which by target cells became dependent on peptide receptor binding. When nude mice carrying human tumor xenografts were treated with branched NT(8-13)-methotrexate, a 60% reduction in tumor growth was observed with respect to mice treated with the free drug. [Mol Cancer Ther 2007;6(9):2441–8]


Protein Science | 2010

Effect of ligand binding on human d-amino acid oxidase: Implications for the development of new drugs for schizophrenia treatment

Laura Caldinelli; Gianluca Molla; Luisa Bracci; Barbara Lelli; Silvia Pileri; Pamela Cappelletti; Silvia Sacchi; Loredano Pollegioni

In human brain the flavoprotein D‐amino acid oxidase (hDAAO) is responsible for the degradation of the neuromodulator D‐serine, an important effector of NMDA‐receptor mediated neurotransmission. Experimental evidence supports the concept that D‐serine concentration increase by hDAAO inhibition may represent a valuable therapeutic approach to improve the symptoms in schizophrenia patients. This study investigated the effects on hDAAO conformation and stability of the substrate D‐serine (or of the pseudo‐substrate trifluoro‐D‐alanine), the FAD cofactor, and two inhibitors (benzoate, a classical substrate‐competitive inhibitor and the drug chlorpromazine (CPZ), which competes with the cofactor). We demonstrated that all these compounds do not alter the interaction of hDAAO with its physiological partner pLG72. The ligands used affect the tertiary structure of hDAAO differently: benzoate or trifluoro‐D‐alanine binding increases the amount of the holoenzyme form in solution and stabilizes the flavoprotein, while CPZ binding favors a protein conformation resembling that of the apoprotein, which is more sensitive to degradation. Interestingly, the apoprotein form of hDAAO binds the substrate D‐serine: this interaction increases FAD binding thus increasing the amount of active holoenzyme in solution. Benzoate and CPZ similarly modify the short‐term cellular D‐serine concentration but affect the cellular concentration of hDAAO differently. In conclusion, the different alteration of hDAAO conformation and stability by the ligands used represents a further parameter to take into consideration during the development of new drugs to cope schizophrenia.


ChemMedChem | 2011

Target-Selective Drug Delivery through Liposomes Labeled with Oligobranched Neurotensin Peptides

Chiara Falciani; Antonella Accardo; Jlenia Brunetti; Diego Tesauro; Barbara Lelli; Alessandro Pini; Luisa Bracci; Giancarlo Morelli

The structure and the in vitro behavior of liposomes filled with the cytotoxic drug doxorubicin (Doxo) and functionalized on the external surface with a branched moiety containing four copies of the 8–13 neurotensin (NT) peptide is reported. The new functionalized liposomes, DOPC‐NT4Lys(C18)2, are obtained by co‐aggregation of the DOPC phospholipid with a new synthetic amphiphilic molecule, NT4Lys(C18)2, which contains a lysine scaffold derivatized with a lipophilic moiety and a tetrabranched hydrophilic peptide, NT8–13, a neurotensin peptide fragment well known for its ability to mimic the neurotensin peptide in receptor binding ability. Dynamic light scattering measurements indicate a value for the hydrodynamic radius (RH) of 88.3±4.4 nm. The selective internalization and cytotoxicity of DOPC‐NT4Lys(C18)2 liposomes containing Doxo, as compared to pure DOPC liposomes, were tested in HT29 human colon adenocarcinoma and TE671 human rhabdomyosarcoma cells, both of which express neurotensin receptors. Peptide‐functionalized liposomes show a clear advantage in comparison to pure DOPC liposomes with regard to drug internalization in both HT29 and TE671 tumor cells: FACS analysis indicates an increase in fluorescence signal of the NT4‐liposomes, compared to the DOPC pure analogues, in both cell lines; cytotoxicity of DOPC‐NT4Lys(C18)2‐Doxo liposomes is increased four‐fold with respect to DOPC‐Doxo liposomes in both HT29 and TE671 cell lines. These effects could to be ascribed to the higher rate of internalization for DOPC‐NT4Lys(C18)2‐Doxo liposomes, due to stronger binding driven by a lower dissociation constant of the NT4‐liposomes that bind the membrane onto a specific protein, in contrast to DOPC liposomes, which approach the plasma membrane unselectively.


PLOS ONE | 2012

Isomerization of an Antimicrobial Peptide Broadens Antimicrobial Spectrum to Gram-Positive Bacterial Pathogens

Chiara Falciani; Luisa Lozzi; Simona Pollini; Vincenzo Luca; Veronica Carnicelli; Jlenia Brunetti; Barbara Lelli; Stefano Bindi; Silvia Scali; Antonio Di Giulio; Gian Maria Rossolini; Maria Luisa Mangoni; Luisa Bracci; Alessandro Pini

The branched M33 antimicrobial peptide was previously shown to be very active against Gram-negative bacterial pathogens, including multidrug-resistant strains. In an attempt to produce back-up molecules, we synthesized an M33 peptide isomer consisting of D-aminoacids (M33-D). This isomeric version showed 4 to 16-fold higher activity against Gram-positive pathogens, including Staphylococcus aureus and Staphylococcus epidermidis, than the original peptide, while retaining strong activity against Gram-negative bacteria. The antimicrobial activity of both peptides was influenced by their differential sensitivity to bacterial proteases. The better activity shown by M33-D against S. aureus compared to M33-L was confirmed in biofilm eradication experiments where M33-L showed 12% activity with respect to M33-D, and in vivo models where Balb-c mice infected with S. aureus showed 100% and 0% survival when treated with M33-D and M33-L, respectively. M33-D appears to be an interesting candidate for the development of novel broad-spectrum antimicrobials active against bacterial pathogens of clinical importance.


Biochemical Journal | 2006

Stable peptide inhibitors prevent binding of lethal and oedema factors to protective antigen and neutralize anthrax toxin in vivo

Alessandro Pini; Ylenia Runci; Chiara Falciani; Barbara Lelli; Jlenia Brunetti; Silvia Pileri; Monica Fabbrini; Luisa Lozzi; Claudia Ricci; Andrea Bernini; Fiorella Tonello; Federica Dal Molin; Paolo Neri; Neri Niccolai; Luisa Bracci

The lethal and oedema toxins produced by Bacillus anthracis, the aetiological agent of anthrax, are made by association of protective antigen with lethal and oedema factors and play a major role in the pathogenesis of anthrax. In the present paper, we describe the production of peptide-based specific inhibitors in branched form which inhibit the interaction of protective antigen with lethal and oedema factors and neutralize anthrax toxins in vitro and in vivo. Anti-protective antigen peptides were selected from a phage library by competitive panning with lethal factor. Selected 12-mer peptides were synthesized in tetra-branched form and were systematically modified to obtain peptides with higher affinity and inhibitory efficiency.


ChemMedChem | 2010

Design and in vitro evaluation of branched peptide conjugates:turning nonspecific cytotoxic drugs into tumor-selective agents.

Chiara Falciani; Jlenia Brunetti; Chiara Pagliuca; Stefano Menichetti; Lucia Vitellozzi; Barbara Lelli; Alessandro Pini; Luisa Bracci

The use of peptide receptors as targets for tumor‐selective therapies was envisaged years ago with the findings that receptors for different endogenous regulatory peptides are overexpressed in several primary and metastatic human tumors, and can be used as tumor antigens. Branched peptides can retain or even increase, through multivalent binding, the biological activity of a peptide and are very resistant to proteolysis, thus having a markedly higher in vivo activity compared with the corresponding monomeric peptides. Oligo‐branched peptides, containing the human regulatory peptide neurotensin (NT) sequence, have been used as tumor‐specific targeting agents. These peptides are able to selectively and specifically deliver effector units, for cell imaging or killing, to tumor cells that overexpress NT receptors. Results obtained with branched NT conjugated to different functional units for tumor imaging and therapy indicate that branched peptides are promising novel multifunctional targeting molecules. This study is focused on the role of the releasing pattern of drug‐conjugated branched NT peptides. We present results obtained with oligo‐branched neurotensin peptides conjugated to 6‐mercaptopurin (6‐MP), combretastain A‐4 (CA4) and monastrol (MON). Drugs were conjugated to oligo‐branched neurotensin through different linkers, and the mode‐of‐release, together with cytotoxicity, was studied in different human cancer cell lines. The results show that branched peptides are very promising pharmacodelivery options. Among our drug‐armed branched peptides, NT4–CA4 was identified as a candidate for further development and evaluation in preclinical pharmacokinetic and pharmacodynamic studies. This peptide–drug exhibits significant activity against pancreas and prostate human cancer cells. Consequently, this derivative is of considerable interest due to the high mortality rates of pancreas neuroendocrine tumors and the high incidence of prostate cancer.


Current Cancer Drug Targets | 2010

Modular branched neurotensin peptides for tumor target tracing and receptor-mediated therapy: a proof-of-concept.

Chiara Falciani; Barbara Lelli; Jlenia Brunetti; Silvia Pileri; Alessandra Cappelli; Alessandro Pini; Chiara Pagliuca; Niccolò Ravenni; Lapo Bencini; Stefano Menichetti; Renato Moretti; M De Prizio; Marco Scatizzi; Luisa Bracci

The aim of this study was to demonstrate that oligo-branched peptides can be effective either for spotlighting tumor cells that overexpress peptide receptors, or for killing them, simply by exchanging the functional moiety coupled to the conserved receptor-targeting core. Tetra-branched peptides containing neurotensin (NT) sequence are described here as selective targeting agents for human colon, pancreas and prostate cancer. Fluorophore-conjugated peptides were used to measure tumor versus healthy tissue binding in human surgical samples, resulting in validation of neurotensin receptors as highly promising tumor-biomarkers. Drug-armed branched peptides were synthesized with different conjugation methods, resulting in uncleavable adducts or drug-releasing molecules. Cytotoxicity on human cell lines from colon (HT-29), pancreas (PANC-1) or prostate (PC-3) carcinoma indicated branched NT conjugated with MTX and 5-FdU as the most active agents on PANC-1 (EC(50) 4.4e-007 M) and HT-29 (1.1e-007 M), respectively. Tetra-branched NT armed with 5-FdU was used for in vivo experiments in HT-29-xenografted mice and produced a 50% reduction in tumor growth with respect to animals treated with the free drug. An unrelated branched peptide carrying the same drug was completely ineffective. In vitro and in vivo results indicated that branched peptides are valuable tools for tumor selective targeting.


ChemMedChem | 2012

Structure-activity relationships and mechanism of action of Eph-ephrin antagonists: interaction of cholanic acid with the EphA2 receptor

Massimiliano Tognolini; Matteo Incerti; Iftiin Hassan-Mohamed; Carmine Giorgio; Simonetta Russo; Renato Bruni; Barbara Lelli; Luisa Bracci; Roberta Noberini; Elena B. Pasquale; Elisabetta Barocelli; Paola Vicini; Marco Mor; Alessio Lodola

The Eph–ephrin system, including the EphA2 receptor and the ephrinA1 ligand, plays a critical role in tumor and vascular functions during carcinogenesis. We previously identified (3α,5β)‐3‐hydroxycholan‐24‐oic acid (lithocholic acid) as an Eph–ephrin antagonist that is able to inhibit EphA2 receptor activation; it is therefore potentially useful as a novel EphA2 receptor‐targeting agent. Herein we explore the structure–activity relationships of a focused set of lithocholic acid derivatives based on molecular modeling investigations and displacement binding assays. Our exploration shows that while the 3‐α‐hydroxy group of lithocholic acid has a negligible role in recognition of the EphA2 receptor, its carboxylate group is critical for disrupting the binding of ephrinA1 to EphA2. As a result of our investigation, we identified (5β)‐cholan‐24‐oic acid (cholanic acid) as a novel compound that competitively inhibits the EphA2–ephrinA1 interaction with higher potency than lithocholic acid. Surface plasmon resonance analysis indicates that cholanic acid binds specifically and reversibly to the ligand binding domain of EphA2, with a steady‐state dissociation constant (KD) in the low micromolar range. Furthermore, cholanic acid blocks the phosphorylation of EphA2 as well as cell retraction and rounding in PC3 prostate cancer cells, two effects that depend on EphA2 activation by the ephrinA1 ligand. These findings suggest that cholanic acid can be used as a template structure for the design of effective EphA2 antagonists, and may have potential impact in the elucidation of the role played by this receptor in pathological conditions.

Collaboration


Dive into the Barbara Lelli'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
Researchain Logo
Decentralizing Knowledge