1
|
Basile S, Parisi C, Bellia F, Zimbone S, Arrabito G, Gulli D, Pignataro B, Giuffrida ML, Sortino S, Copani A. Red-Light-Photosensitized Tyrosine 10 Nitration of β-Amyloid 1-42 Diverts the Protein from Forming Toxic Aggregates. ACS Chem Neurosci 2024; 15:2916-2924. [PMID: 39036818 DOI: 10.1021/acschemneuro.4c00284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Several studies have highlighted the presence of nitration damage following neuroinflammation in Alzheimer's disease (AD). Accordingly, post-transcriptional modifications of β-amyloid (Aβ), including peptide nitration, have been explored as a marker of the disease. However, the implications of Aβ nitration in terms of aggregation propensity and neurotoxicity are still debated. Here, we show new data obtained using a photoactivatable peroxynitrite generator (BPT-NO) to overcome the limitations associated with chemical nitration methods. We found that the photoactivation of BPT-NO with the highly biocompatible red light selectively induces the nitration of tyrosine 10 of freshly solubilized full-length Aβ1-42. Photonitrated Aβ1-42 was, therefore, investigated for aggregation states and functions. It resulted that photonitrated Aβ1-42 did not aggregate into small oligomers but rather self-assembled into large amorphous aggregates. When tested on neuronal-like SH-SY5Y cells and microglial C57BL/6 BV2 cells, photonitrated Aβ1-42 showed to be free of neurotoxicity and able to induce phagocytic microglia cells. We propose that light-controlled nitration of the multiple forms in which Aβ occurs (i.e., monomers, oligomers, fibrils) could be a tool to assess in real-time the impact of tyrosine nitration on the amyloidogenic and toxic properties of Aβ1-42.
Collapse
Affiliation(s)
- Sarah Basile
- Department of Drug and Health Sciences (DSFS), University of Catania, 95125 Catania, Italy
| | - Cristina Parisi
- Department of Drug and Health Sciences (DSFS), University of Catania, 95125 Catania, Italy
| | - Francesco Bellia
- Institute of Crystallography (IC), National Research Council, 95126 Catania, Italy
| | - Stefania Zimbone
- Institute of Crystallography (IC), National Research Council, 95126 Catania, Italy
| | - Giuseppe Arrabito
- Department of Physics and Chemistry (DiFC) Emilio Segrè, University of Palermo, 90128 Palermo, Italy
- ATeN Center, University of Palermo, 90128 Palermo, Italy
| | - Daniele Gulli
- ATeN Center, University of Palermo, 90128 Palermo, Italy
| | - Bruno Pignataro
- Department of Physics and Chemistry (DiFC) Emilio Segrè, University of Palermo, 90128 Palermo, Italy
- ATeN Center, University of Palermo, 90128 Palermo, Italy
| | | | - Salvatore Sortino
- Department of Drug and Health Sciences (DSFS), University of Catania, 95125 Catania, Italy
| | - Agata Copani
- Department of Drug and Health Sciences (DSFS), University of Catania, 95125 Catania, Italy
- Institute of Crystallography (IC), National Research Council, 95126 Catania, Italy
| |
Collapse
|
2
|
Martínez-Camarena Á, Bellia F, Paz Clares M, Vecchio G, Nicolas J, García-España E. Polymeric Nanozyme with SOD Activity Capable of Inhibiting Self- and Metal-Induced α-Synuclein Aggregation. Chemistry 2024; 30:e202401331. [PMID: 38687026 DOI: 10.1002/chem.202401331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/02/2024]
Abstract
Despite decades of research, Parkinson's disease is still an idiopathic pathology for which no cure has yet been found. This is partly explained by the multifactorial character of most neurodegenerative syndromes, whose generation involves multiple pathogenic factors. In Parkinson's disease, two of the most important ones are the aggregation of α-synuclein and oxidative stress. In this work, we address both issues by synthesizing a multifunctional nanozyme based on grafting a pyridinophane ligand that can strongly coordinate CuII, onto biodegradable PEGylated polyester nanoparticles. The resulting nanozyme exhibits remarkable superoxide dismutase activity together with the ability to inhibit the self-induced aggregation of α-synuclein into amyloid-type fibrils. Furthermore, the combination of the chelator and the polymer produces a cooperative effect whereby the resulting nanozyme can also halve CuII-induced α-synuclein aggregation.
Collapse
Affiliation(s)
- Álvaro Martínez-Camarena
- ICMol, Departament de Química Inorgànica, Universitat de València, C/Catedrático José Beltrán 2, Paterna, 46980, Spain
- Institut Galien Paris-Saclay, CNRS, Université Paris-Saclay, Orsay, 91400, France
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, Catania, 95125, Italy
- MatMoPol Research Group, Departamento de Química Inorgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Avda. Complutense s/n, Madrid, 28040, Spain
| | - Francesco Bellia
- Istituto di Cristallografia, CNR, P. Gaifami 18, Catania, 95126, Italy
| | - M Paz Clares
- ICMol, Departament de Química Inorgànica, Universitat de València, C/Catedrático José Beltrán 2, Paterna, 46980, Spain
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, Catania, 95125, Italy
| | - Julien Nicolas
- Institut Galien Paris-Saclay, CNRS, Université Paris-Saclay, Orsay, 91400, France
| | - Enrique García-España
- ICMol, Departament de Química Inorgànica, Universitat de València, C/Catedrático José Beltrán 2, Paterna, 46980, Spain
| |
Collapse
|
3
|
Oliveri V. Unveiling the Effects of Copper Ions in the Aggregation of Amyloidogenic Proteins. Molecules 2023; 28:6446. [PMID: 37764220 PMCID: PMC10537474 DOI: 10.3390/molecules28186446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Amyloid diseases have become a global concern due to their increasing prevalence. Transition metals, including copper, can affect the aggregation of the pathological proteins involved in these diseases. Copper ions play vital roles in organisms, but the disruption of their homeostasis can negatively impact neuronal function and contribute to amyloid diseases with toxic protein aggregates, oxidative stress, mitochondrial dysfunction, impaired cellular signaling, inflammation, and cell death. Gaining insight into the imbalance of copper ions and its impact on protein folding and aggregation is crucial for developing focused therapies. This review examines the influence of copper ions on significant amyloid proteins/peptides, offering a comprehensive overview of the current understanding in this field.
Collapse
Affiliation(s)
- Valentina Oliveri
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A Doria 6, 95125 Catania, Italy
| |
Collapse
|
4
|
Queiroz DD, Ribeiro TP, Gonçalves JM, Mattos LMM, Gerhardt E, Freitas J, Palhano FL, Frases S, Pinheiro AS, McCann M, Knox A, Devereux M, Outeiro TF, Pereira MD. A water-soluble manganese(II) octanediaoate/phenanthroline complex acts as an antioxidant and attenuates alpha-synuclein toxicity. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166475. [PMID: 35777688 DOI: 10.1016/j.bbadis.2022.166475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/24/2022]
Abstract
The overproduction of reactive oxygen species (ROS) induces oxidative stress, a well-known process associated with aging and several human pathologies, such as cancer and neurodegenerative diseases. A large number of synthetic compounds have been described as antioxidant enzyme mimics, capable of eliminating ROS and/or reducing oxidative damage. In this study, we investigated the antioxidant activity of a water-soluble 1,10-phenantroline-octanediaoate Mn2+-complex on cells under oxidative stress, and assessed its capacity to attenuate alpha-synuclein (aSyn) toxicity and aggregation, a process associated with increased oxidative stress. This Mn2+-complex exhibited a significant antioxidant potential, reducing intracelular oxidation and increasing oxidative stress resistance in S. cerevisiae cells and in vivo, in G. mellonella, increasing the activity of the intracellular antioxidant enzymes superoxide dismutase and catalase. Strikingly, the Mn2+-complex reduced both aSyn oligomerization and aggregation in human cell cultures and, using NMR and DFT/molecular docking we confirmed its interaction with the C-terminal region of aSyn. In conclusion, the Mn2+-complex appears as an excellent lead for the design of new phenanthroline derivatives as alternative compounds for preventing oxidative damages and oxidative stress - related diseases.
Collapse
Affiliation(s)
- Daniela D Queiroz
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia, Cidade Universitária, Universidade Federal do Rio de Janeiro, Brazil; Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Germany; Rede de Micrologia RJ-FAPERJ, Brazil
| | - Thales P Ribeiro
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia, Cidade Universitária, Universidade Federal do Rio de Janeiro, Brazil; Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Germany; Rede de Micrologia RJ-FAPERJ, Brazil
| | - Julliana M Gonçalves
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia, Cidade Universitária, Universidade Federal do Rio de Janeiro, Brazil; Rede de Micrologia RJ-FAPERJ, Brazil
| | - Larissa M M Mattos
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia, Cidade Universitária, Universidade Federal do Rio de Janeiro, Brazil; Rede de Micrologia RJ-FAPERJ, Brazil
| | - Ellen Gerhardt
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Germany
| | - Júlia Freitas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando L Palhano
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susana Frases
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Anderson S Pinheiro
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia, Cidade Universitária, Universidade Federal do Rio de Janeiro, Brazil
| | - Malachy McCann
- Department of Chemistry, Maynooth University, Maynooth, Ireland
| | - Andrew Knox
- The Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Camden Row, Dublin 8, Ireland
| | - Michael Devereux
- The Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Camden Row, Dublin 8, Ireland
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Germany; Max Planck Institute for Experimental Medicine, Göttingen, Germany; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK; Scientific employee with an honorary contract at German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
| | - Marcos D Pereira
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia, Cidade Universitária, Universidade Federal do Rio de Janeiro, Brazil; Rede de Micrologia RJ-FAPERJ, Brazil.
| |
Collapse
|
5
|
Wiesen T, Atlas D. Novel anti-apoptotic L-DOPA precursors SuperDopa and SuperDopamide as potential neuroprotective agents for halting/delaying progression of Parkinson’s disease. Cell Death Dis 2022; 13:227. [PMID: 35277478 PMCID: PMC8917195 DOI: 10.1038/s41419-022-04667-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 01/23/2022] [Accepted: 02/07/2022] [Indexed: 11/20/2022]
Abstract
Parkinson’s disease (PD) is characterized by a gradual degeneration of the dopaminergic neurons in the substantia nigra pars compacta (SNpC). Levodopa, the standard PD treatment, provides the missing dopamine in SNpC, but ultimately after a honeymoon with levodopa treatment the neurodegenerative process and the progression of the disease continue. Aimed at prolonging the life of dopaminergic cells, we prepared the levodopa precursors SuperDopa (SD) and SueprDopamide (SDA), in which levodopa is merged with the antioxidant N-acetylcysteine (NAC) into a single molecule. Rotenone is a mitochondrial complex inhibitor often used as experimental model of PD. In vivo, SD and SDA treatment show a significant relief of motor disabilities in rotenone-injected rats. SD and SDA also lower rotenone-induced-α-synuclein (α-syn) expression in human SH-SY5Y cells, and α-syn oligomerization in α-syn-overexpressing-HEK293 cells. In the neuronal SH-SY5Y cells, SD and SDA reverse oxidative stress-induced phosphorylation of cJun-N-terminal kinase (JNK) and p38-mitogen-activated kinase (p38MAPK). Attenuation of the MAPK-inflammatory/apoptotic pathway in SH-SY5Y cells concurrent with protection of rotenone-triggered motor impairment in rats, is a manifestation of the combined antioxidant/anti-inflammatory activity of SD and SDA together with levodopa release. The concept of joined therapies into a single molecule, where levodopa precursors confer antioxidant activity by enabling NAC delivery across the BBB, provides a potential disease-modifying treatment for slowing PD progression.
Collapse
|
6
|
In Vitro Antibacterial, Anti-Adhesive and Anti-Biofilm Activities of Krameria lappacea (Dombey) Burdet & B.B. Simpson Root Extract against Methicillin-Resistant Staphylococcus aureus Strains. Antibiotics (Basel) 2021; 10:antibiotics10040428. [PMID: 33924336 PMCID: PMC8069196 DOI: 10.3390/antibiotics10040428] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 12/12/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a serious threat to public health, due to its large variety of pathogenetic mechanisms. Accordingly, the present study aimed to investigate the anti-MRSA activities of Krameria lappacea, a medicinal plant native to South America. Through Ultra-High-Performance Liquid Chromatography coupled with High-Resolution Mass spectrometry, we analyzed the chemical composition of Krameria lappacea root extract (KLRE). The antibacterial activity of KLRE was determined by the broth microdilution method, also including the minimum biofilm inhibitory concentration and minimum biofilm eradication concentration. Besides, we evaluated the effect on adhesion and invasion of human lung carcinoma A549 cell line by MRSA strains. The obtained results revealed an interesting antimicrobial action of this extract, which efficiently inhibit the growth, biofilm formation, adhesion and invasion of MRSA strains. Furthermore, the chemical analysis revealed the presence in the extract of several flavonoid compounds and type-A and type-B proanthocyanidins, which are known for their anti-adhesive effects. Taken together, our findings showed an interesting antimicrobial activity of KLRE, giving an important contribution to the current knowledge on the biological activities of this plant.
Collapse
|
7
|
Khoramjouy M, Naderi N, Kobarfard F, Heidarli E, Faizi M. An Intensified Acrolein Exposure Can Affect Memory and Cognition in Rat. Neurotox Res 2021; 39:277-291. [PMID: 32876917 DOI: 10.1007/s12640-020-00278-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022]
Abstract
Acrolein is a clear, colorless liquid and a highly reactive α, β-unsaturated aldehyde. Acrolein, a byproduct and initiator of oxidative stress, has a major role in the pathogenesis of disorders including pulmonary, cardiovascular, atherosclerosis, and neurodegenerative diseases. Environmental or dietary exposure and endogenous production are common sources of acrolein. Widespread exposure to acrolein is a major risk for human health; therefore, we decided to investigate the neurological effects of acrolein. In this study, we used male Sprague-Dawley rats and exposed them orally to acrolein (0.5, 1, 3, and 5 mg/kg/day) for 90 days and investigated the neurobehavioral and electrophysiological disturbances. We also assessed the correlation between neurotoxicity and CSF concentration of acrolein in the rats. The results showed that chronic oral administration of acrolein at 5 mg/kg/day impaired learning and memory in the neurobehavioral tests. In addition, acrolein decreased the release of excitatory neurotransmitters such as glutamate in electrophysiological studies. Our data demonstrated that chronic oral exposure of acrolein at a dose of 5 mg/kg leads to a direct correlation between neurotoxicity and its CSF concentration. In conclusion, exposure to acrolein as a major pollutant in the environment may cause cognitive problems and may have serious neurocognitive effects on humans.
Collapse
Affiliation(s)
- Mona Khoramjouy
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Nima Naderi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Farzad Kobarfard
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elmira Heidarli
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Mehrdad Faizi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran.
| |
Collapse
|
8
|
Bellia F, Grasso GI, Ahmed IMM, Oliveri V, Vecchio G. Carnoquinolines Target Copper Dyshomeostasis, Aberrant Protein-Protein Interactions, and Oxidative Stress. Chemistry 2020; 26:16690-16705. [PMID: 32627921 DOI: 10.1002/chem.202001591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 12/20/2022]
Abstract
Metal dysregulation, oxidative stress, protein modification, and aggregation are factors strictly interrelated and associated with neurodegenerative pathologies. As such, all of these aspects represent valid targets to counteract neurodegeneration and, therefore, the development of metal-binding compounds with other properties to combat multifactorial disorders is definitely on the rise. Herein, the synthesis and in-depth analysis of the first hybrids of carnosine and 8-hydroxyquinoline, carnoquinolines (CarHQs), which combine the properties of the dipeptide with those of 8-hydroxyquinoline, are reported. CarHQs and their copper complexes were characterized through several techniques, such as ESI-MS and NMR, UV/Vis, and circular dichroism spectroscopy. CarHQs can modulate self- and copper-induced amyloid-β aggregation. These hybrids combine the antioxidant activity of their parent compounds. Therefore, they can simultaneously scavenge free radicals and reactive carbonyl species, thanks to the phenolic group and imidazole ring. These results indicate that CarHQs are promising multifunctional candidates for neurodegenerative disorders and they are worthy of further studies.
Collapse
Affiliation(s)
- Francesco Bellia
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, P. Gaifami 18, 95126, Catania, Italy
| | - Giuseppa Ida Grasso
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125, Catania, Italy
| | | | - Valentina Oliveri
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
9
|
|
10
|
Sbardella D, Coletta A, Tundo GR, Ahmed IMM, Bellia F, Oddone F, Manni G, Coletta M. Structural and functional evidence for citicoline binding and modulation of 20S proteasome activity: Novel insights into its pro-proteostatic effect. Biochem Pharmacol 2020; 177:113977. [PMID: 32298691 DOI: 10.1016/j.bcp.2020.113977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/10/2020] [Indexed: 02/01/2023]
Abstract
Citicoline or CDP-choline is a drug, made up by a cytidine 5'-diphosphate moiety and choline, which upon adsorption is rapidly hydrolyzed into cytidine 5'-diphosphate and choline, easily bypassing the blood-brain barrier. Once in the brain, these metabolites are used to re-synthesize citicoline in neurons and in the other cell histo-types which uptake them. Citicoline administration finds broad therapeutic application in the treatment of glaucoma as well as other retinal disorders by virtue of its safety profile and neuro-protective and neuroenhancer activity, which significantly improves the visual function. Further, though supported by limited clinical studies, this molecule finds therapeutic application in neurodegenerative disease, delaying the cognitive decline in Alzheimer's Disease (AD) and Parkinson's Disease (PD) subjects. In this work we show that citicoline greatly affects the proteolytic activity of the 20S proteasome on synthetic and natural substrates, functioning as a bimodal allosteric modulator, likely binding at multiple sites. In silico binding simulations identify several potential binding sites for citicoline on 20S proteasome, and their topology envisages the possibility that, by occupying some of these pockets, citicoline may induce a conformational shift of the 20S proteasome, allowing to sketch a working hypothesis for the structural basis of its function as allosteric modulator. In addition, we show that over the same concentration range citicoline affects the distribution of assembled proteasome populations and turn-over of ubiquitinated proteins in SH-SY5Y and SK-N-BE human neuroblastoma cells, suggesting its potential role as a regulator of proteostasis in nervous cells.
Collapse
Affiliation(s)
- Diego Sbardella
- IRCCS-Fondazione G.B. BIETTI, via Livenza, 3, 00189, Rome, Italy.
| | - Andrea Coletta
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Grazia Raffaella Tundo
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, via Montpellier, 1, 00133, Rome, Italy
| | - Ikhlas M M Ahmed
- Institute of Crystallography, CNR, via Gaifami, 18, 95126, Catania, Italy
| | - Francesco Bellia
- Institute of Crystallography, CNR, via Gaifami, 18, 95126, Catania, Italy
| | - Francesco Oddone
- IRCCS-Fondazione G.B. BIETTI, via Livenza, 3, 00189, Rome, Italy
| | - Gianluca Manni
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, via Montpellier, 1, 00133, Rome, Italy
| | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, via Montpellier, 1, 00133, Rome, Italy.
| |
Collapse
|