1
|
Cosottini L, Geri A, Ghini V, Mannelli M, Zineddu S, Di Paco G, Giachetti A, Massai L, Severi M, Gamberi T, Rosato A, Turano P, Messori L. Unlocking the Power of Human Ferritin: Enhanced Drug Delivery of Aurothiomalate in A2780 Ovarian Cancer Cells. Angew Chem Int Ed Engl 2024; 63:e202410791. [PMID: 38949226 DOI: 10.1002/anie.202410791] [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: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/02/2024]
Abstract
Aurothiomalate (AuTM) is an FDA-approved antiarthritic gold drug with unique anticancer properties. To enhance its anticancer activity, we prepared a bioconjugate with human apoferritin (HuHf) by attaching some AuTM moieties to surface protein residues. The reaction of apoferritin with excess AuTM yielded a single adduct, that was characterized by ESI MS and ICP-OES analysis, using three mutant ferritins and trypsinization experiments. The adduct contains ~3 gold atoms per ferritin subunit, arranged in a small cluster bound to Cys90 and Cys102. MD simulations provided a plausible structural model for the cluster. The adduct was evaluated for its pharmacological properties and was found to be significantly more cytotoxic than free AuTM against A2780 cancer cells mainly due to higher gold uptake. NMR-metabolomics showed that AuTM bound to HuHf and free AuTM induced qualitatively similar changes in treated cancer cells, indicating that the effects on cell metabolism are approximately the same, in agreement with independent biochemical experiments. In conclusion, we have demonstrated here that a molecularly precise bioconjugate formed between AuTM and HuHf exhibits anticancer properties far superior to the free drug, while retaining its key mechanistic features. Evidence is provided that human ferritin can serve as an excellent carrier for this metallodrug.
Collapse
Affiliation(s)
- Lucrezia Cosottini
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Andrea Geri
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Veronica Ghini
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Stefano Zineddu
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Giorgio Di Paco
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Andrea Giachetti
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), 50019, Sesto Fiorentino, FI, Italy
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Mirko Severi
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Antonio Rosato
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
- Magnetic Resonance Center, University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Paola Turano
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
- Magnetic Resonance Center, University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| |
Collapse
|
2
|
Ceccherini V, Giorgi E, Mannelli M, Cirri D, Gamberi T, Gabbiani C, Pratesi A. Synthesis, Chemical Characterization, and Biological Evaluation of Hydrophilic Gold(I) and Silver(I) N-Heterocyclic Carbenes as Potential Anticancer Agents. Inorg Chem 2024; 63:16949-16963. [PMID: 39226133 DOI: 10.1021/acs.inorgchem.4c02581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
A series of new gold(I) and silver(I) N-heterocyclic carbenes bearing a 1-thio-β-d-glucose tetraacetate moiety was synthesized and chemically characterized. The compounds' stability and solubility in physiological conditions were investigated employing a multitechnique approach. Interaction studies with biologically relevant proteins, such as superoxide dismutase (SOD) and human serum albumin (HSA), were conducted via UV-vis absorption spectroscopy and high-resolution ESI mass spectrometry. The biological activity of the compounds was evaluated in the A2780 and A2780R (cisplatin-resistant) ovarian cancer cell lines and the HSkMC (human skeletal muscle) healthy cell line. Inhibition studies of the selenoenzyme thioredoxin reductase (TrxR) were also carried out. The results highlighted that the gold complexes are more stable in aqueous environment and capable of interaction with SOD and HSA. Moreover, these carbenes strongly inhibited the TrxR activity. In contrast, the silver ones underwent structural alterations in the aqueous medium and showed greater antiproliferative activity.
Collapse
Affiliation(s)
- Valentina Ceccherini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Ester Giorgi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| |
Collapse
|
3
|
Galassi R, Sargentoni N, Renzi S, Luciani L, Bartolacci C, Pattabhi P, Andreani C, Pucciarelli S. Anticancer Activity of Imidazolyl Gold(I/III) Compounds in Non-Small Cell Lung Cancer Cell Lines. Pharmaceuticals (Basel) 2024; 17:1133. [PMID: 39338298 PMCID: PMC11435220 DOI: 10.3390/ph17091133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/31/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Lung cancer is a leading cause of cancer-related death worldwide that needs updated therapies to contrast both the serious side effects and the occurrence of drug resistance. A panel of non-small cell lung cancer (NSCLC) cells were herein employed as cancer models. Eight structurally related gold(I) and gold(III) complexes with NHC and halides or triphenylphosphane ligands were investigated as lung cancer cell growth inhibitors. As expected, gold compounds with PPh3 were found to be more cytotoxic than homoleptic [(NHC)2-Au(I)]X or heteroleptic NHC-Au(I)X or NHC-Au(III)X3 complexes. Mixed ligand gold(I) compounds exhibiting the linear NHC-AuPPh3 (compound 7) or the trigonal NHC-Au(Cl)PPh3 (compound 8) arrangements at the central metal were found to be the best lung cancer cytotoxic compounds. Analysis of the TrxR residual activity of the treated cells revealed that these compounds efficiently inhibit the most accredited molecular target for gold compounds, the TrxR, with compound 8 reaching more than 80% activity reduction in lung cells. Some of the current cancer lung therapy protocols consist of specific lung cancer cell cytotoxic agents combined with antifolate drugs; interestingly, the herein gold compounds are both TrxR and antifolate inhibitors. The human DHFR was inhibited with IC50 ranging between 10-21 µM, depending on substrate concentrations, proceeding by a likely allosteric mechanism only for compound 8.
Collapse
Affiliation(s)
- Rossana Galassi
- Chemistry Division, School of Science and Technology, University of Camerino, ChIP Via Madonna delle Carceri, 62032 Camerino, Italy; (N.S.); (L.L.)
| | - Nicola Sargentoni
- Chemistry Division, School of Science and Technology, University of Camerino, ChIP Via Madonna delle Carceri, 62032 Camerino, Italy; (N.S.); (L.L.)
| | - Sofia Renzi
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (S.R.); (S.P.)
| | - Lorenzo Luciani
- Chemistry Division, School of Science and Technology, University of Camerino, ChIP Via Madonna delle Carceri, 62032 Camerino, Italy; (N.S.); (L.L.)
| | - Caterina Bartolacci
- University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45219, USA; (C.B.); (P.P.); (C.A.)
| | - Prasad Pattabhi
- University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45219, USA; (C.B.); (P.P.); (C.A.)
| | - Cristina Andreani
- University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45219, USA; (C.B.); (P.P.); (C.A.)
| | - Stefania Pucciarelli
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy; (S.R.); (S.P.)
| |
Collapse
|
4
|
Vitali V, Massai L, Messori L. Strategies for the design of analogs of auranofin endowed with anticancer potential. Expert Opin Drug Discov 2024; 19:855-867. [PMID: 38803122 DOI: 10.1080/17460441.2024.2355329] [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: 03/06/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Auranofin (AF) is a well-established, FDA-approved, antiarthritic gold drug that is currently being reevaluated for a variety of therapeutic indications through drug repurposing. AF has shown great promise as a potential anticancer agent and has been approved for a few clinical trials in cancer. The renewed interest in AF has led to extensive research into the design, preparation and biological evaluation of auranofin analogs, which may have an even better pharmacological profile than the parent drug. AREAS COVERED This article reviews the strategies for chemical modification of the AF scaffold. Several auranofin analogs have been prepared and characterized for medical application in the field of cancer treatment over the last 20 years. Some emerging structure-function relationships are proposed and discussed. EXPERT OPINION The chemical modification of the AF scaffold has been the subject of intense activity in recent years and this strategy has led to the preparation and evaluation of several AF analogs. The case of iodauranofin is a particularly promising example. The availability of homogeneous biological data for a group of AF derivatives allows some initial structure-function relationships to be proposed, which may inspire the design and synthesis of new and better AF analogs for cancer treatment.
Collapse
Affiliation(s)
- Valentina Vitali
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Lara Massai
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
5
|
Bernabeu De Maria M, Matczuk M, Tesauro D, Saviano M, Sikorski J, Chiappetta G, Godin S, Szpunar J, Lobinski R, Ronga L. Study of metalation of thioredoxin by gold(I) therapeutic compounds using combined liquid chromatography/capillary electrophoresis with inductively coupled plasma/electrospray MS/MS detection. Anal Bioanal Chem 2024; 416:2819-2833. [PMID: 38244050 DOI: 10.1007/s00216-024-05140-z] [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: 11/16/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
The reactivity of thioredoxin (Trx1) with the Au(I) drug auranofin (AF) and two therapeutic N-heterocyclic carbene (NHC)2-Au(I) complexes (bis [1-methyl-3-acridineimidazolin-2-ylidene]gold(I) tetrafluoroborate (Au3BC) and [1,3-diethyl-4,5-bis(4methoxyphenyl)imidazol-2-ylidene]gold(I) (Au4BC)) was investigated. Direct infusion (DI) electrospray ionization (ESI) mass spectrometry (MS) allowed information on the structure, stoichiometry, and kinetics of formation of Trx-Au adducts. The fragmentation of the formed adducts in the gas phase gave insights into the exact Au binding site within the protein, demonstrating the preference for Trx1 Cys32 or Cys35 of AF or the (NHC)2-Au(I) complex Au3BC, respectively. Reversed-phase HPLC suffered from the difficulty of elution of gold compounds, did not preserve the formed metal-protein adducts, and favored the loss of ligands (phosphine or NHC) from Au(I). These limitations were eliminated by capillary electrophoresis (CE) which enabled the separation of the gold compounds, Trx1, and the formed adducts. The ICP-MS/MS detection allowed the simultaneous quantitative monitoring of the gold and sulfur isotopes and the determination of the metallation extent of the protein. The hyphenation of the mentioned techniques was used for the analysis of Trx1-Au adducts for the first time.
Collapse
Affiliation(s)
- Mikel Bernabeu De Maria
- Université de Pau Et Des Pays de L'Adour, E2S UPPA, CNRS, Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM-UMR 5254), 64053, Pau, France
| | - Magdalena Matczuk
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664, Warsaw, Poland
| | - Diego Tesauro
- Department of Pharmacy and CIRPeB, Università degli Studi di Napoli Federico II, Via Montesano 49, 80131, Naples, Italy
| | - Michele Saviano
- Instituto Di Cristallografia (IC), CNR, 70126, Caserta, Italy
| | - Jacek Sikorski
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664, Warsaw, Poland
| | - Giovanni Chiappetta
- Biological Mass Spectrometry and Proteomics (SMBP), ESPCI Paris, Université PSL, LPC CNRS UMR8249, 75005, Paris, France
| | - Simon Godin
- Université de Pau Et Des Pays de L'Adour, E2S UPPA, CNRS, Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM-UMR 5254), 64053, Pau, France
| | - Joanna Szpunar
- Université de Pau Et Des Pays de L'Adour, E2S UPPA, CNRS, Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM-UMR 5254), 64053, Pau, France
| | - Ryszard Lobinski
- Université de Pau Et Des Pays de L'Adour, E2S UPPA, CNRS, Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM-UMR 5254), 64053, Pau, France
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664, Warsaw, Poland
| | - Luisa Ronga
- Université de Pau Et Des Pays de L'Adour, E2S UPPA, CNRS, Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM-UMR 5254), 64053, Pau, France.
| |
Collapse
|
6
|
Geri A, Zineddu S, Massai L, Ronga L, Lobinski R, Gailer J, Messori L. Mercury binding to proteins disclosed by ESI MS experiments: The case of three organomercurials. J Inorg Biochem 2024; 252:112479. [PMID: 38218139 DOI: 10.1016/j.jinorgbio.2024.112479] [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: 10/12/2023] [Revised: 12/01/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024]
Abstract
Solution interactions of three organomercury compounds, i.e., methylmercury chloride, thimerosal and phenylmercury acetate, with a group of biochemically relevant proteins, namely cytochrome c (Cyt c), ribonuclease A (RNase A), carbonic anhydrase I (hCA I), superoxide dismutase (SOD), and serum albumin (HSA), were investigated using an established ESI MS approach. Temporal analysis of sample aliquots provided insight into the binding kinetics, while comparative analysis of the obtained mass spectra disclosed adduct formation of each mercurial with the tested proteins and the relative abundance of the species. The three organomercurials bind, exclusively and tightly, to free cysteine residues as no binding was observed in the case of proteins lacking such groups. hCA I, SOD and HSA formed distinct mercury adducts, preserving the Hg bound alkyl/aryl ligands; yet, the three organomercurials displayed significant differences in reactivity in relation to their chemical structure. The investigation was then extended to analyze the reactions with the C-terminal dodecapeptide of the enzyme human thioredoxin reductase, which contains a characteristic selenol-thiol moiety: tight Hg binding was observed. Notably, this peptide was able to remove effectively and completely the alkyl/aryl ligands of the three tested organomercurials; this behavior may be relevant to the detoxification mechanism of organomercurials in mammals. Finally, a competition experiment was carried out to establish whether protein bound mercury centers may be displaced by other competing metals. Interestingly, and quite unexpectedly, we observed that a protein bound mercury fragment may be partially displaced from its coordination site in hCA I by the medicinal gold compound auranofin.
Collapse
Affiliation(s)
- Andrea Geri
- Laboratory of Metals in Medicine, Department of Chemistry, University of Florence, via della Lastruccia 3, 50019, Sesto Fiorentino, Florence, Italy
| | - Stefano Zineddu
- Laboratory of Metals in Medicine, Department of Chemistry, University of Florence, via della Lastruccia 3, 50019, Sesto Fiorentino, Florence, Italy
| | - Lara Massai
- Laboratory of Metals in Medicine, Department of Chemistry, University of Florence, via della Lastruccia 3, 50019, Sesto Fiorentino, Florence, Italy.
| | - Luisa Ronga
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Pau, France
| | - Ryszard Lobinski
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Pau, France; Chair of Analytical Chemistry, Warsaw University of Technology, ul.Noakowskiego 3, 00-664 Warszawa, Poland
| | - Jürgen Gailer
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Luigi Messori
- Laboratory of Metals in Medicine, Department of Chemistry, University of Florence, via della Lastruccia 3, 50019, Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
7
|
Giorgi E, Mannelli M, Gamberi T, Durante M, Gabbiani C, Cirri D, Pratesi A. Cytotoxic auranofin analogues bearing phosphine, arsine and stibine ligands: A study on the possible role of the ligand on the biological activity. J Inorg Biochem 2024; 251:112452. [PMID: 38070433 DOI: 10.1016/j.jinorgbio.2023.112452] [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: 09/25/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/25/2023]
Abstract
Three gold(I) linear compounds, sharing the general formula [AuI(LPh3)], have been synthesized and characterized. The nature of the ligand has been modified by moving down among some of the elements of group 15, i.e. phosphorus, arsenic and antimony. The structures of derived compounds have been solved through XRD and the reactivity behaviour towards selected biomolecules has been investigated through a multi-technique approach involving NMR, high-resolution mass spectrometry and IR. Moreover, the biological activity of the investigated compounds has been comparatively analyzed through classical methodologies and the disclosed differences are discussed in detail.
Collapse
Affiliation(s)
- Ester Giorgi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Maria Durante
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy.
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy.
| |
Collapse
|
8
|
Geri A, Massai L, Novinec M, Turel I, Messori L. Reactions of Medicinal Gold Compounds with Cathepsin B Explored through Electrospray Mass Spectrometry Measurements. Chempluschem 2024; 89:e202300321. [PMID: 37930642 DOI: 10.1002/cplu.202300321] [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: 07/03/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Medicinal gold compounds, a novel class of potential anticancer drugs, are believed to produce their pharmacological effects mainly through direct gold binding to protein targets at the level of solvent exposed cysteine (or selenocysteine) residues. We have explored therein the reactions of a panel of seven representative gold compounds with the cysteine protease cathepsin B according to an established ESI MS approach. Detailed information on the mode of protein binding of these gold compounds is gained; notably, quite distinct patterns of cathepsin B metalation have emerged from these studies. It is shown that panel gold compounds interact preferentially, often exclusively, with the free cysteine located in the active site of the enzyme.
Collapse
Affiliation(s)
- Andrea Geri
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Marko Novinec
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, 1000, Ljubljana, Slovenia
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, 1000, Ljubljana, Slovenia
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
9
|
Coverdale JPC, Polepalli S, Arruda MAZ, da Silva ABS, Stewart AJ, Blindauer CA. Recent Advances in Metalloproteomics. Biomolecules 2024; 14:104. [PMID: 38254704 PMCID: PMC10813065 DOI: 10.3390/biom14010104] [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: 10/20/2023] [Revised: 11/17/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Interactions between proteins and metal ions and their complexes are important in many areas of the life sciences, including physiology, medicine, and toxicology. Despite the involvement of essential elements in all major processes necessary for sustaining life, metalloproteomes remain ill-defined. This is not only owing to the complexity of metalloproteomes, but also to the non-covalent character of the complexes that most essential metals form, which complicates analysis. Similar issues may also be encountered for some toxic metals. The review discusses recently developed approaches and current challenges for the study of interactions involving entire (sub-)proteomes with such labile metal ions. In the second part, transition metals from the fourth and fifth periods are examined, most of which are xenobiotic and also tend to form more stable and/or inert complexes. A large research area in this respect concerns metallodrug-protein interactions. Particular attention is paid to separation approaches, as these need to be adapted to the reactivity of the metal under consideration.
Collapse
Affiliation(s)
- James P. C. Coverdale
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Edgbaston B15 2TT, UK;
| | | | - Marco A. Z. Arruda
- Institute of Chemistry, Department of Analytical Chemistry, Universidade Estadual de Campinas, Campinas 13083-970, Brazil; (M.A.Z.A.); (A.B.S.d.S.)
| | - Ana B. Santos da Silva
- Institute of Chemistry, Department of Analytical Chemistry, Universidade Estadual de Campinas, Campinas 13083-970, Brazil; (M.A.Z.A.); (A.B.S.d.S.)
| | - Alan J. Stewart
- School of Medicine, University of St. Andrews, St Andrews KY16 9TF, UK
| | | |
Collapse
|
10
|
Chiaverini L, Baglini E, Mannelli M, Poggetti V, Da Settimo F, Taliani S, Gamberi T, Barresi E, La Mendola D, Marzo T. A complex bearing TSPO PIGA ligand coordinated to the [Au(PEt 3)] + pharmacophore is highly cytotoxic against ovarian cancer cells. Biometals 2023; 36:961-968. [PMID: 36869967 PMCID: PMC10545567 DOI: 10.1007/s10534-023-00496-8] [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: 12/19/2022] [Accepted: 02/09/2023] [Indexed: 03/05/2023]
Abstract
Auranofin ([1-(thio-κS)-β-D-glucopyranose-2,3,4,6-tetraacetato](triethylphosphine)-gold) is a leading gold-based drug clinically used to treat arthritis. In the last years, it entered various drug reprofiling programs, and it has been found promising against various forms of tumor, including ovarian cancer. Evidence showed as its antiproliferative profile mainly depends on the inhibition of thioredoxin reductase (TrxR), being this mitochondrial system its main target. In this context, we report here the synthesis and biological evaluation of a novel complex designed as auranofin analogue obtained through the conjugation of a phenylindolylglyoxylamide ligand (which belongs to the so-called PIGA TSPO ligand family) with the auranofin-derived cationic fragment [Au(PEt3)]+. This complex is characterized by two parts. The phenylindolylglyoxylamide moiety, owing to its high affinity for TSPO (in the low nM range) should drive the compound to target mitochondria, whereas the [Au(PEt3)]+ cation is the actual anticancer-active molecular fragment. Overall, we wanted to offer the proof-of-concept that by coupling PIGA ligands to anticancer gold active moieties, it is possible to preserve and even improve anticancer effects, opening the avenue to a reliable approach for targeted therapy.
Collapse
Affiliation(s)
- Lorenzo Chiaverini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy.
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy.
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
- University Consortium for Research in the Chemistry of Metal Ions in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126, Bari, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
- University Consortium for Research in the Chemistry of Metal Ions in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126, Bari, Italy
| |
Collapse
|
11
|
Yan W, Zhong Y, Hu X, Xu T, Zhang Y, Kales S, Qu Y, Talley DC, Baljinnyam B, LeClair CA, Simeonov A, Polster BM, Huang R, Ye Y, Rai G, Henderson MJ, Tao D, Fang S. Auranofin targets UBA1 and enhances UBA1 activity by facilitating ubiquitin trans-thioesterification to E2 ubiquitin-conjugating enzymes. Nat Commun 2023; 14:4798. [PMID: 37558718 PMCID: PMC10412574 DOI: 10.1038/s41467-023-40537-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023] Open
Abstract
UBA1 is the primary E1 ubiquitin-activating enzyme responsible for generation of activated ubiquitin required for ubiquitination, a process that regulates stability and function of numerous proteins. Decreased or insufficient ubiquitination can cause or drive aging and many diseases. Therefore, a small-molecule enhancing UBA1 activity could have broad therapeutic potential. Here we report that auranofin, a drug approved for the treatment of rheumatoid arthritis, is a potent UBA1 activity enhancer. Auranofin binds to the UBA1's ubiquitin fold domain and conjugates to Cys1039 residue. The binding enhances UBA1 interactions with at least 20 different E2 ubiquitin-conjugating enzymes, facilitating ubiquitin charging to E2 and increasing the activities of seven representative E3s in vitro. Auranofin promotes ubiquitination and degradation of misfolded ER proteins during ER-associated degradation in cells at low nanomolar concentrations. It also facilitates outer mitochondrial membrane-associated degradation. These findings suggest that auranofin can serve as a much-needed tool for UBA1 research and therapeutic exploration.
Collapse
Affiliation(s)
- Wenjing Yan
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yongwang Zhong
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Xin Hu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Tuan Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yinghua Zhang
- Center for Innovative Biomedical Resources, Biosensor Core, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Stephen Kales
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yanyan Qu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Daniel C Talley
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Bolormaa Baljinnyam
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Christopher A LeClair
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA.
| | - Shengyun Fang
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Program in Oncology, UM Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
12
|
Geri A, Massai L, Messori L. Protein Metalation by Medicinal Gold Compounds: Identification of the Main Features of the Metalation Process through ESI MS Experiments. Molecules 2023; 28:5196. [PMID: 37446857 DOI: 10.3390/molecules28135196] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Gold compounds form a new class of promising anticancer agents with innovative modes of action. It is generally believed that anticancer gold compounds, at variance with clinically established platinum drugs, preferentially target proteins rather than nucleic acids. The reactions of several gold compounds with a few model proteins have been systematically explored in recent years through ESI MS measurements to reveal adduct formation and identify the main features of those reactions. Here, we focus our attention on a group of five gold compounds of remarkable medicinal interest, i.e., Auranofin, Au(NHC)Cl, [Au(NHC)2]PF6, Aubipyc, and Auoxo6, and on their reactions with four different biomolecular targets, i.e., the proteins HEWL, hCA I, HSA and the C-terminal dodecapeptide of the enzyme thioredoxin reductase. Complete ESI MS data are available for those reactions due to previous experimental work conducted in our laboratory. From the comparative analysis of the ESI MS reaction profiles, some characteristic trends in the metallodrug-protein reactivity may be identified as detailed below. The main features are described and analyzed in this review. Overall, all these observations are broadly consistent with the concept that cytotoxic gold drugs preferentially target cancer cell proteins, with a remarkable selectivity for the cysteine and selenocysteine proteome. These interactions typically result in severe damage to cancer cell metabolism and profound alterations in the redox state, leading to eventual cancer cell death.
Collapse
Affiliation(s)
- Andrea Geri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Florence, Italy
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Florence, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Florence, Italy
| |
Collapse
|
13
|
Ronga L, Tolbatov I, Giorgi E, Pisarek P, Enjalbal C, Marrone A, Tesauro D, Lobinski R, Marzo T, Cirri D, Pratesi A. Mechanistic Evaluations of the Effects of Auranofin Triethylphosphine Replacement with a Trimethylphosphite Moiety. Inorg Chem 2023; 62:10389-10396. [PMID: 37342994 PMCID: PMC10324304 DOI: 10.1021/acs.inorgchem.3c01280] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 06/23/2023]
Abstract
Auranofin, a gold(I)-based complex, is under clinical trials for application as an anticancer agent for the treatment of nonsmall-cell lung cancer and ovarian cancer. In the past years, different derivatives have been developed, modifying gold linear ligands in the search for new gold complexes endowed with a better pharmacological profile. Recently, a panel of four gold(I) complexes, inspired by the clinically established compound auranofin, was reported by our research group. As described, all compounds possess an [Au{P(OMe)3}]+ cationic moiety, in which the triethylphosphine of the parent compound auranofin was replaced with an oxygen-rich trimethylphosphite ligand. The gold(I) linear coordination geometry was complemented by Cl-, Br-, I-, and the auranofin-like thioglucose tetraacetate ligand. As previously reported, despite their close similarity to auranofin, the panel compounds exhibited some peculiar and distinctive features, such as lower log P values which can induce relevant differences in the overall pharmacokinetic profiles. To get better insight into the P-Au strength and stability, an extensive study was carried out for relevant biological models, including three different vasopressin peptide analogues and cysteine, using 31P NMR and LC-ESI-MS. A DFT computational study was also carried out for a better understanding of the theoretical fundamentals of the disclosed differences with regard to triethylphosphine parent compounds.
Collapse
Affiliation(s)
- Luisa Ronga
- Université
de Pau et des Pays de l’Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
| | - Iogann Tolbatov
- Institute
of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Av. Paisos Catalans 16, 43007 Tarragona, Spain
| | - Ester Giorgi
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Paulina Pisarek
- Université
de Pau et des Pays de l’Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
| | - Christine Enjalbal
- IBMM,
Université de Montpellier, CNRS, ENSCM, UMR 5247, 34293 Montpellier, France
| | - Alessandro Marrone
- Department
of Pharmacy, University “G. D’Annunzio”
Chieti-Pescara, Via dei
Vestini, 31, 66100 Chieti, Italy
| | - Diego Tesauro
- Department
of Pharmacy and CIRPeB, Università
degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Ryszard Lobinski
- Université
de Pau et des Pays de l’Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
- Chair
of Analytical Chemistry, Department of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Tiziano Marzo
- Department
of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy
| | - Damiano Cirri
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Alessandro Pratesi
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| |
Collapse
|
14
|
Gopal J, Muthu M, Sivanesan I. A Comprehensive Survey on the Expediated Anti-COVID-19 Options Enabled by Metal Complexes-Tasks and Trials. Molecules 2023; 28:molecules28083354. [PMID: 37110587 PMCID: PMC10143858 DOI: 10.3390/molecules28083354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Contemporary pharmacology dating back to the late 19th/early 20th centuries has benefitted largely from the incorporation of metal complexes. Various biological attributes have been successfully realized using metal/metal complex-based drugs. Among anticancer, antimicrobial, and antiviral applications, anticancer applications have extracted the maximum benefit from the metal complex, Cisplatin. The following review has compiled the various antiviral benefits harnessed through inputs from metal complexes. As a result of exploiting the pharmacological aspects of metal complexes, the anti-COVID-19 deliverables have been summarized. The challenges ahead, the gaps in this research area, the need to improvise incorporating nanoaspects in metal complexes, and the need to test metal complex-based drugs in clinical trials have been discussed and deliberated. The pandemic shook the entire world and claimed quite a percentage of the global population. Metal complex-based drugs are already established for their antiviral property with respect to enveloped viruses and extrapolating them for COVID-19 can be an effective way to manipulate drug resistance and mutant issues that the current anti-COVID-19 drugs are facing.
Collapse
Affiliation(s)
- Judy Gopal
- Department of Research and Innovation, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Manikandan Muthu
- Department of Research and Innovation, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Iyyakkannu Sivanesan
- Department of Bioresources and Food Science, Institute of Natural Science and Agriculture, Konkuk University, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
15
|
Chemical Modification of Auranofin Yields a New Family of Anticancer Drug Candidates: The Gold(I) Phosphite Analogues. Molecules 2023; 28:molecules28031050. [PMID: 36770719 PMCID: PMC9920260 DOI: 10.3390/molecules28031050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
A panel of four novel gold(I) complexes, inspired by the clinically established gold drug auranofin (1-Thio-β-D-glucopyranosatotriethylphosphine gold-2,3,4,6-tetraacetate), was prepared and characterized. All these compounds feature the replacement of the triethylphosphine ligand of the parent compound auranofin with a trimethylphosphite ligand. The linear coordination around the gold(I) center is completed by Cl-, Br-, I- or by the thioglucose tetraacetate ligand (SAtg). The in-solution behavior of these gold compounds as well as their interactions with some representative model proteins were comparatively analyzed through 31PNMR and ESI-MS measurements. Notably, all panel compounds turned out to be stable in aqueous media, but significant differences with respect to auranofin were disclosed in their interactions with a few leading proteins. In addition, the cytotoxic effects produced by the panel compounds toward A2780, A2780R and SKOV-3 ovarian cancer cells were quantitated and found to be in the low micromolar range, since the IC50 of all compounds was found to be between 1 μM and 10 μM. Notably, these novel gold complexes showed large and similar inhibition capabilities towards the key enzyme thioredoxin reductase, again comparable to those of auranofin. The implications of these results for the discovery of new and effective gold-based anticancer agents are discussed.
Collapse
|
16
|
Giampà M, Corinti D, Maccelli A, Fornarini S, Berden G, Oomens J, Schwarzbich S, Glaser T, Crestoni ME. Binding Modes of a Cytotoxic Dinuclear Copper(II) Complex with Phosphate Ligands Probed by Vibrational Photodissociation Ion Spectroscopy. Inorg Chem 2023; 62:1341-1353. [PMID: 36655890 PMCID: PMC9890465 DOI: 10.1021/acs.inorgchem.2c02091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The dinuclear copper complex bearing a 2,7-disubstituted-1,8-naphthalenediol ligand, [(HtomMe){Cu(OAc)}2](OAc), a potential anticancer drug able to bind to two neighboring phosphates in the DNA backbone, is endowed with stronger cytotoxic effects and inhibition ability of DNA synthesis in human cancer cells as compared to cisplatin. In this study, the intrinsic binding ability of the charged complex [(HtomMe){Cu(OAc)}2]+ is investigated with representative phosphate diester ligands with growing chemical complexity, ranging from simple inorganic phosphate up to mononucleotides. An integrated method based on high-resolution mass spectrometry (MS), tandem MS, and infrared multiple photon dissociation (IRMPD) spectroscopy in the 600-1800 cm-1 spectral range, backed by quantum chemical calculations, has been used to characterize complexes formed in solution and delivered as bare species by electrospray ionization. The structural features revealed by IRMPD spectroscopy have been interpreted by comparison with linear IR spectra of the lowest-energy structures, revealing diagnostic signatures of binding modes of the dinuclear copper(II) complex with phosphate groups, whereas the possible competitive interaction with the nucleobase is silenced in the gas phase. This result points to the prevailing interaction of [(HtomMe){Cu(OAc)}2]+ with phosphate diesters and mononucleotides as a conceivable contribution to the observed anticancer activity.
Collapse
Affiliation(s)
- Marco Giampà
- Department
of Clinical and Molecular Medicine, Norwegian
University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway
| | - Davide Corinti
- Dipartimento
di Chimica e Tecnologie del Farmaco, Università
di Roma “La Sapienza”, I-00185 Roma, Italy,
| | - Alessandro Maccelli
- Dipartimento
di Chimica e Tecnologie del Farmaco, Università
di Roma “La Sapienza”, I-00185 Roma, Italy
| | - Simonetta Fornarini
- Dipartimento
di Chimica e Tecnologie del Farmaco, Università
di Roma “La Sapienza”, I-00185 Roma, Italy
| | - Giel Berden
- Institute
for Molecules and Materials, FELIX Laboratory, Radboud University, Toernooiveld 7, 6525 ED Nijmegen, The Netherlands
| | - Jos Oomens
- Institute
for Molecules and Materials, FELIX Laboratory, Radboud University, Toernooiveld 7, 6525 ED Nijmegen, The Netherlands
| | - Sabrina Schwarzbich
- Lehrstuhl
für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, D-33615 Bielefeld, Germany
| | - Thorsten Glaser
- Lehrstuhl
für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, D-33615 Bielefeld, Germany
| | - Maria Elisa Crestoni
- Dipartimento
di Chimica e Tecnologie del Farmaco, Università
di Roma “La Sapienza”, I-00185 Roma, Italy,
| |
Collapse
|
17
|
Binacchi F, Elia C, Cirri D, Van de Griend C, Zhou XQ, Messori L, Bonnet S, Pratesi A, Biver T. A biophysical study of the interactions of palladium(II), platinum(II) and gold(III) complexes of aminopyridyl-2,2'-bipyridine ligands with RNAs and other nucleic acid structures. Dalton Trans 2023; 52:598-608. [PMID: 36562298 DOI: 10.1039/d2dt03483b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Metal compounds form an attractive class of ligands for a variety of nucleic acids. Five metal complexes bearing aminopyridyl-2,2'-bipyridine tetradentate ligands and possessing a quasi-planar geometry were challenged toward different types of nucleic acid molecules including RNA polynucleotides in the duplex or triplex form, an RNA Holliday four-way junction, natural double helix DNA and a DNA G-quadruplex. The binding process was monitored comparatively using different spectroscopic and melting methods. The binding preferences that emerge from our analysis are discussed in relation to the structural features of the metal complexes.
Collapse
Affiliation(s)
- Francesca Binacchi
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, 56124 Pisa, Italy.
| | - Cassandra Elia
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, 56124 Pisa, Italy.
| | - Damiano Cirri
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, 56124 Pisa, Italy.
| | - Corjan Van de Griend
- Leiden Institute of Chemistry, Leiden University, 2333CC Leiden, The Netherlands
| | - Xue-Quan Zhou
- Leiden Institute of Chemistry, Leiden University, 2333CC Leiden, The Netherlands
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University, 2333CC Leiden, The Netherlands
| | - Alessandro Pratesi
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, 56124 Pisa, Italy.
| | - Tarita Biver
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, 56124 Pisa, Italy.
| |
Collapse
|
18
|
Selenol (-SeH) as a target for mercury and gold in biological systems: Contributions of mass spectrometry and atomic spectroscopy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
19
|
Milutinović MG, Milivojević NN, Đorđević NM, Nikodijević DD, Radisavljević SR, Đeković Kesić AS, Marković SD. Gold(III) Complexes with Phenanthroline-derivatives Ligands Induce Apoptosis in Human Colorectal and Breast Cancer Cell Lines. J Pharm Sci 2022; 111:3215-3223. [PMID: 36162493 DOI: 10.1016/j.xphs.2022.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 01/05/2023]
Abstract
Due to their promising effects, gold(III) complexes recently drew increasing attention in the design of new metal-based anticancer therapeutics. Two gold(III) complexes, square-planar [Au(DPP)Cl2]+ - Complex 1 and distorted square-pyramidal [Au(DMP)Cl3] - Complex 2 (where DPP=4,7-diphenyl-1,10-phenanthroline and DMP=2,9-dimethyl-1,10-phenanthroline) were previously synthetized, described and approved as complexes with pronounced cytotoxic effects on colorectal HCT-116 and breast MDA-MB-231 cancer cells. This study investigated the type of cell death by AO/EB double staining, and identification of possible targets responsible for their cytotoxicity, monitored by immunofluorescence and qPCR methods. Both complexes induced apoptosis in all applied concentrations. In the HCT-116 cells apoptosis was activated by external apoptotic pathway, via increase of Fas receptor protein expression and Caspase 8 gene expression. Also, the mitochondrial pathway was triggered by affecting the Bcl-2 members of regulatory proteins and increased caspase 9 protein expression. In MDA-MB-231 cells, apoptosis was initiated from the mitochondria, due to disbalance between expressions of pro- and anti-apoptotic Bcl-2 family members and caspase 9 activation. Complex 1 shows better activity compared to Complex 2, which is in accordance with its structural characteristics. The results deal weighty data about proapoptotic activity of gold(III) complexes and highlighted potential targets for cancer therapy.
Collapse
Affiliation(s)
- Milena G Milutinović
- University of Kragujevac, Department of Biology and Ecology, Faculty of Science, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Nevena N Milivojević
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Natural Sciences, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Nevena M Đorđević
- University of Kragujevac, Department of Biology and Ecology, Faculty of Science, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Danijela D Nikodijević
- University of Kragujevac, Department of Biology and Ecology, Faculty of Science, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Snežana R Radisavljević
- University of Kragujevac, Department of Chemistry, Faculty of Science, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Ana S Đeković Kesić
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Natural Sciences, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Snežana D Marković
- University of Kragujevac, Department of Biology and Ecology, Faculty of Science, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| |
Collapse
|
20
|
Evaluation of Auranofin Loading within Ferritin Nanocages. Int J Mol Sci 2022; 23:ijms232214162. [PMID: 36430642 PMCID: PMC9695178 DOI: 10.3390/ijms232214162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/02/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Auranofin (AF), a gold(I) compound that is currently used for the treatment of rheumatoid arthritis and is in clinical trials for its promising anticancer activity, was encapsulated within the human H-chain and the horse spleen ferritin nanocages using the alkaline disassembly/reassembly protocol. The aim of the work was to highlight possible differences in their drug loading capacity and efficacy. The drug-loaded ferritins were characterized via UV-vis absorption spectroscopy and inductively coupled plasma-atomic emission spectroscopy to assess AF encapsulation and to define the exact amount of gold atoms trapped in the Ft cavity. The crystal structures allowed us to define the nature of AF interaction with both ferritins and to identify the gold binding sites. Moreover, the biological characterization let us to obtain preliminary information on the cytotoxic effect of AF when bound to the human H-chain.
Collapse
|
21
|
Massai L, Grifagni D, De Santis A, Geri A, Cantini F, Calderone V, Banci L, Messori L. Gold-Based Metal Drugs as Inhibitors of Coronavirus Proteins: The Inhibition of SARS-CoV-2 Main Protease by Auranofin and Its Analogs. Biomolecules 2022; 12:1675. [PMID: 36421689 PMCID: PMC9687241 DOI: 10.3390/biom12111675] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 08/08/2023] Open
Abstract
Gold compounds have a long tradition in medicine and offer many opportunities for new therapeutic applications. Herein, we evaluated the lead compound Auranofin and five related gold(I) complexes as possible inhibitors of SARS-CoV-2 Main Protease (SARS-CoV-2 Mpro), a validated drug target for the COVID-19 disease. The investigational panel of gold compounds included Auranofin; three halido analogues, i.e., Au(PEt3)Cl, Au(PEt3)Br, and Au(PEt3)I; and two gold carbene complexes, i.e., Au(NHC)Cl and [Au(NHC)2]PF6. Notably, all these gold compounds, with the only exception of [Au(NHC)2]PF6, turned out to be potent inhibitors of the catalytic activity of SARS-CoV-2 Mpro: the measured Ki values were in the range 2.1-0.4 μM. The reactions of the various gold compounds with SARS-CoV-2 Mpro were subsequently investigated through electrospray ionization (ESI) mass spectrometry (MS) upon a careful optimization of the experimental conditions; the ESI MS spectra provided clear evidence for the formation of tight metallodrug-protein adducts and for the coordination of well defined gold-containing fragments to the SARS-CoV-2 Mpro, again with the only exception of [Au(NHC)2]PF6, The metal-protein stoichiometry was unambiguously determined for the resulting species. The crystal structures of the metallodrug- Mpro adducts were solved in the case of Au(PEt3)Br and Au(NHC)Cl. These crystal structures show that gold coordination occurs at the level of catalytic Cys 145 in the case of Au(NHC)Cl and at the level of both Cys 145 and Cys 156 for Au(PEt3)Br. Tight coordination of gold atoms to functionally relevant cysteine residues is believed to represent the true molecular basis of strong enzyme inhibition.
Collapse
Affiliation(s)
- Lara Massai
- Department of Chemistry “Ugo Schiff”, University of Florence, Via Della Lastruccia 3, 50019 Florence, Italy
| | - Deborah Grifagni
- Department of Chemistry “Ugo Schiff”, University of Florence, Via Della Lastruccia 3, 50019 Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Alessia De Santis
- Department of Chemistry “Ugo Schiff”, University of Florence, Via Della Lastruccia 3, 50019 Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Andrea Geri
- Department of Chemistry “Ugo Schiff”, University of Florence, Via Della Lastruccia 3, 50019 Florence, Italy
| | - Francesca Cantini
- Department of Chemistry “Ugo Schiff”, University of Florence, Via Della Lastruccia 3, 50019 Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), University of Florence, Via L. Sacconi 6, 50019 Florence, Italy
| | - Vito Calderone
- Department of Chemistry “Ugo Schiff”, University of Florence, Via Della Lastruccia 3, 50019 Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), University of Florence, Via L. Sacconi 6, 50019 Florence, Italy
| | - Lucia Banci
- Department of Chemistry “Ugo Schiff”, University of Florence, Via Della Lastruccia 3, 50019 Florence, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), University of Florence, Via L. Sacconi 6, 50019 Florence, Italy
| | - Luigi Messori
- Department of Chemistry “Ugo Schiff”, University of Florence, Via Della Lastruccia 3, 50019 Florence, Italy
| |
Collapse
|
22
|
Russo L, Giacomelli C, Fortino M, Marzo T, Ferri G, Calvello M, Viegi A, Magrì A, Pratesi A, Pietropaolo A, Cardarelli F, Martini C, Rizzarelli E, Marchetti L, La Mendola D, Trincavelli ML. Neurotrophic Activity and Its Modulation by Zinc Ion of a Dimeric Peptide Mimicking the Brain-Derived Neurotrophic Factor N-Terminal Region. ACS Chem Neurosci 2022; 13:3453-3463. [PMID: 36346920 PMCID: PMC9732821 DOI: 10.1021/acschemneuro.2c00463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin (NT) essential for neuronal development and synaptic plasticity. Dysregulation of BDNF signaling is implicated in different neurological disorders. The direct NT administration as therapeutics has revealed to be challenging. This has prompted the design of peptides mimicking different regions of the BDNF structure. Although loops 2 and 4 have been thoroughly investigated, less is known regarding the BDNF N-terminal region, which is involved in the selective recognition of the TrkB receptor. Herein, a dimeric form of the linear peptide encompassing the 1-12 residues of the BDNF N-terminal (d-bdnf) was synthesized. It demonstrated to act as an agonist promoting specific phosphorylation of TrkB and downstream ERK and AKT effectors. The ability to promote TrkB dimerization was investigated by advanced fluorescence microscopy and molecular dynamics (MD) simulations, finding activation modes shared with BDNF. Furthermore, d-bdnf was able to sustain neurite outgrowth and increase the expression of differentiation (NEFM, LAMC1) and polarization markers (MAP2, MAPT) demonstrating its neurotrophic activity. As TrkB activity is affected by zinc ions in the synaptic cleft, we first verified the ability of d-bdnf to coordinate zinc and then the effect of such complexation on its activity. The d-bdnf neurotrophic activity was reduced by zinc complexation, demonstrating the role of the latter in tuning the activity of the new peptido-mimetic. Taken together our data uncover the neurotrophic properties of a novel BDNF mimetic peptide and pave the way for future studies to understand the pharmacological basis of d-bdnf action and develop novel BDNF-based therapeutic strategies.
Collapse
Affiliation(s)
- Lara Russo
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy
| | | | | | - Tiziano Marzo
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy
| | - Gianmarco Ferri
- Laboratorio
NEST, Scuola Normale Superiore, Pisa 56127, Italy
| | | | | | - Antonio Magrì
- Istituto
di Cristallografia, Consiglio Nazionale delle Ricerche (CNR), Catania 95126, Italy
| | - Alessandro Pratesi
- Dipartimento
di Chimica e Chimica Industriale, Università
di Pisa, Pisa 56124, Italy
| | | | | | - Claudia Martini
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy
| | - Enrico Rizzarelli
- Istituto
di Cristallografia, Consiglio Nazionale delle Ricerche (CNR), Catania 95126, Italy,Università
degli Studi di Catania, Catania 95124, Italy
| | - Laura Marchetti
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy,
| | - Diego La Mendola
- Dipartimento
di Farmacia, Università di Pisa, Pisa 56127, Italy,
| | | |
Collapse
|
23
|
Speciation Analysis Highlights the Interactions of Auranofin with the Cytoskeleton Proteins of Lung Cancer Cells. Pharmaceuticals (Basel) 2022; 15:ph15101285. [PMID: 36297397 PMCID: PMC9610265 DOI: 10.3390/ph15101285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 12/01/2022] Open
Abstract
Two types of lung cells (epithelial cancer lung cells, A-549 and lung fibroblasts MRC-5) were exposed to the clinically established gold drug auranofin at concentrations close to the half-maximal inhibitory drug concentrations (IC50). Collected cells were subjected to speciation analysis using inductively coupled plasma mass spectrometry (ICP-MS). Auranofin showed better affinity toward proteins than DNA, RNA, and hydrophilic small molecular weight compounds. It can bind to proteins that vary in size (~20 kDa, ~75 kDa, and ≥200 kDa) and pI. However, the possibility of dimerization and protein–protein complex formation should also be taken into account. µRPLC/CZE-ESI-MS/MS studies on trypsinized proteins allowed the indication of 76 peptides for which signal intensity was influenced by auranofin presence in cells. Based on it, identity was proposed for 20 proteins. Except for thioredoxin reductase (TrxR), which is directly targeted by gold complex, the proteins were found to be transformed. Five indicated proteins: myosin, plectin, talin, two annexins, and kinase M3K5, are responsible for cell–cell, cell–protein interactions, and cell motility. A wound healing test confirmed their regulation by auranofin as cell migration decreased by 40% while the cell cycle was not interrupted.
Collapse
|
24
|
Cirri D, Massai L, Giacomelli C, Trincavelli ML, Guerri A, Gabbiani C, Messori L, Pratesi A. Synthesis, chemical characterization, and biological evaluation of a novel auranofin derivative as an anticancer agent. Dalton Trans 2022; 51:13527-13539. [PMID: 36000524 DOI: 10.1039/d2dt00836j] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel gold(I) complex inspired by the known medicinal inorganic compounds auranofin and thimerosal, namely ethylthiosalicylate(triethylphosphine)gold(I) (AFETT hereafter), was synthesized and characterised and its structure was resolved through X-ray diffraction. The solution behavior of AFETT and its interactions with two biologically relevant proteins (i.e. human serum albumin and haemoglobin) and with a synthetic dodecapeptide reproducing the C-terminal portion of thioredoxin reductase were comparatively analyzed through 31P NMR and ESI-MS. Remarkable binding properties toward these biomolecules were disclosed. Moreover, the cytotoxic effects produced by AFETT on two ovarian cancer cell lines (A2780 and A2780 R) and one colorectal cancer cell line (HCT116) were analyzed and found to be strong and nearly superimposable to those of auranofin. Interestingly, for both compounds, the ability to induce downregulation of vimentin expression in A2780 R cells was evidenced. Despite its close similarity to auranofin, AFETT is reported to exhibit some peculiar and distinctive features such as a lower lipophilicity, an increased water solubility and a faster reactivity towards the selected target biomolecules. These differences might confer to AFETT significant pharmaceutical and therapeutic advantages over auranofin itself.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy.
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Chiara Giacomelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | | | - Annalisa Guerri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy.
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy.
| |
Collapse
|
25
|
Abate C, Carnamucio F, Giuffrè O, Foti C. Metal-Based Compounds in Antiviral Therapy. Biomolecules 2022; 12:933. [PMID: 35883489 PMCID: PMC9312833 DOI: 10.3390/biom12070933] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 11/16/2022] Open
Abstract
In recent years, the study of metal complexes and metal-based nanomaterials has aroused particular interest, leading to the promotion of new effective systems for the abatement of various viral diseases. Starting from the analysis of chemical properties, this review focuses on the employment of metal-based nanoparticles as antiviral drugs and how this interaction leads to a substantial enhancement in antiviral activity. The use of metal-based antiviral drugs has also spread for the formulation of antiviral vaccines, thanks especially to the remarkable adjuvant activities of some of the metal complexes. In particular, the small size and inert nature of Au- and Ag-based nanoparticles have been exploited for the design of systems for antiviral drug delivery, leading to the development of specific and safe therapies that lead to a decrease in side effects.
Collapse
Affiliation(s)
| | | | - Ottavia Giuffrè
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy; (C.A.); (F.C.); (C.F.)
| | | |
Collapse
|
26
|
Massai L, Messori L, Carpentieri A, Amoresano A, Melchiorre C, Fiaschi T, Modesti A, Gamberi T, Magherini F. The effects of two gold-N-heterocyclic carbene (NHC) complexes in ovarian cancer cells: a redox proteomic study. Cancer Chemother Pharmacol 2022; 89:809-823. [PMID: 35543764 PMCID: PMC9135895 DOI: 10.1007/s00280-022-04438-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/15/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE Ovarian cancer is the fifth leading cause of cancer-related deaths in women. Standard treatment consists of tumor debulking surgery followed by platinum and paclitaxel chemotherapy; yet, despite the initial response, about 70-75% of patients develop resistance to chemotherapy. Gold compounds represent a family of very promising anticancer drugs. Among them, we previously investigated the cytotoxic and pro-apoptotic properties of Au(NHC) and Au(NHC)2PF6, i.e., a monocarbene gold(I) complex and the corresponding bis(carbene) complex. Gold compounds are known to alter the redox state of cells interacting with free cysteine and selenocysteine residues of several proteins. Herein, a redox proteomic study has been carried out to elucidate the mechanisms of cytotoxicity in A2780 human ovarian cancer cells. METHODS A biotinylated iodoacetamide labeling method coupled with mass spectrometry was used to identify oxidation-sensitive protein cysteines. RESULTS Gold carbene complexes cause extensive oxidation of several cellular proteins; many affected proteins belong to two major functional classes: carbohydrate metabolism, and cytoskeleton organization/cell adhesion. Among the affected proteins, Glyceraldehyde-3-phosphate dehydrogenase inhibition was proved by enzymatic assays and by ESI-MS studies. We also found that Au(NHC)2PF6 inhibits mitochondrial respiration impairing complex I function. Concerning the oxidized cytoskeletal proteins, gold binding to the free cysteines of actin was demonstrated by ESI-MS analysis. Notably, both gold compounds affected cell migration and invasion. CONCLUSIONS In this study, we deepened the mode of action of Au(NHC) and Au(NHC)2PF6, identifying common cellular targets but confirming their different influence on the mitochondrial function.
Collapse
Affiliation(s)
- Lara Massai
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, Sesto Fiorentino, 50019, Firenze, Italy
| | - Luigi Messori
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, Sesto Fiorentino, 50019, Firenze, Italy
| | - Andrea Carpentieri
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Angela Amoresano
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Chiara Melchiorre
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy.
| | - Francesca Magherini
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
27
|
Cheng XF, Wang N, Jiang Z, Chen Z, Niu Y, Tong L, Yu T, Tang B. Quantitative Chemoproteomic Profiling of Targets of Au(I) Complexes by Competitive Activity-Based Protein Profiling. Bioconjug Chem 2022; 33:1131-1137. [PMID: 35576584 DOI: 10.1021/acs.bioconjchem.2c00080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Owing to the encouraging pharmacological action and acceptable toxicity profile, Au(I) complexes have attracted growing interest in the application of disease treatment. In order to investigate their potential target proteins and related bioinformation, herein, we screened four Au(I) complexes and explored the binding proteins utilizing a competitive activity-based protein profiling (ABPP) strategy, including identification experiments and reactivity classification experiments, which offers a simple and robust method to identify the target proteins of Au(I) complexes. We quantified the target proteins of the four Au(I) complexes and found that most of proteins were associated with cancer. In addition, the newly Au(I)-binding proteins and biological gold-protein interaction pathways were exhibited. Furthermore, we estimated the correlation between target proteins of Au(I) complexes and various cancers, which will promote the development of the gold anticancer drugs.
Collapse
Affiliation(s)
- Xiu-Fen Cheng
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Nan Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Zhongyao Jiang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Zhenzhen Chen
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Yaxin Niu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Lili Tong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Ting Yu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| |
Collapse
|
28
|
Chiaverini L, Pratesi A, Cirri D, Nardinocchi A, Tolbatov I, Marrone A, Di Luca M, Marzo T, La Mendola D. Anti-Staphylococcal Activity of the Auranofin Analogue Bearing Acetylcysteine in Place of the Thiosugar: An Experimental and Theoretical Investigation. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082578. [PMID: 35458776 PMCID: PMC9032686 DOI: 10.3390/molecules27082578] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 11/16/2022]
Abstract
Auranofin (AF, hereafter) is an orally administered chrysotherapeutic agent approved for the treatment of rheumatoid arthritis that is being repurposed for various indications including bacterial infections. Its likely mode of action involves the impairment of the TrxR system through the binding of the pharmacophoric cation [AuPEt3]+. Accordingly, a reliable strategy to expand the medicinal profile of AF is the replacement of the thiosugar moiety with different ligands. Herein, we aimed to prepare the AF analogue bearing the acetylcysteine ligand (AF-AcCys, hereafter) and characterize its anti-staphylococcal activity. Biological studies revealed that AF-AcCys retains an antibacterial effect superimposable with that of AF against Staphylococcus aureus, whereas it is about 20 times less effective against Staphylococcus epidermidis. Bioinorganic studies confirmed that upon incubation with human serum albumin, AF-AcCys, similarly to AF, induced protein metalation through the [AuPEt3]+ fragment. Additionally, AF-AcCys appeared capable of binding the dodecapeptide Ac-SGGDILQSGCUG-NH2, corresponding to the tryptic C-terminal fragment (488–499) of hTrxR. To shed light on the pharmacological differences between AF and AF-AcCys, we carried out a comparative experimental stability study and a theoretical estimation of bond dissociation energies, unveiling the higher strength of the Au–S bond in AF-AcCys. From the results, it emerged that the lower lipophilicity of AF-AcCys with respect to AF could be a key feature for its different antibacterial activity. The differences and similarities between AF and AF-AcCys are discussed, alongside the opportunities and consequences that chemical structure modifications imply.
Collapse
Affiliation(s)
- Lorenzo Chiaverini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy; (L.C.); (D.L.M.)
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via G. Moruzzi, 13, 56124 Pisa, Italy; (A.P.); (D.C.)
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via G. Moruzzi, 13, 56124 Pisa, Italy; (A.P.); (D.C.)
| | - Arianna Nardinocchi
- Department of Biology, University of Pisa, Via San Zeno 35–39, 56100 Pisa, Italy;
| | - Iogann Tolbatov
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, 43007 Tarragona, Spain
- Correspondence: (I.T.); (M.D.L.); (T.M.)
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università degli Studi “G. D’Annunzio” Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy;
| | - Mariagrazia Di Luca
- Department of Biology, University of Pisa, Via San Zeno 35–39, 56100 Pisa, Italy;
- Correspondence: (I.T.); (M.D.L.); (T.M.)
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy; (L.C.); (D.L.M.)
- Correspondence: (I.T.); (M.D.L.); (T.M.)
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy; (L.C.); (D.L.M.)
| |
Collapse
|
29
|
Chiaverini L, Pratesi A, Cirri D, Nardinocchi A, Tolbatov I, Marrone A, Di Luca M, Marzo T, La Mendola D. Anti-Staphylococcal Activity of the Auranofin Analogue Bearing Acetylcysteine in Place of the Thiosugar: An Experimental and Theoretical Investigation. Molecules 2022. [PMID: 35458776 DOI: 10.3390/molecules27082578/s1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Auranofin (AF, hereafter) is an orally administered chrysotherapeutic agent approved for the treatment of rheumatoid arthritis that is being repurposed for various indications including bacterial infections. Its likely mode of action involves the impairment of the TrxR system through the binding of the pharmacophoric cation [AuPEt3]+. Accordingly, a reliable strategy to expand the medicinal profile of AF is the replacement of the thiosugar moiety with different ligands. Herein, we aimed to prepare the AF analogue bearing the acetylcysteine ligand (AF-AcCys, hereafter) and characterize its anti-staphylococcal activity. Biological studies revealed that AF-AcCys retains an antibacterial effect superimposable with that of AF against Staphylococcus aureus, whereas it is about 20 times less effective against Staphylococcus epidermidis. Bioinorganic studies confirmed that upon incubation with human serum albumin, AF-AcCys, similarly to AF, induced protein metalation through the [AuPEt3]+ fragment. Additionally, AF-AcCys appeared capable of binding the dodecapeptide Ac-SGGDILQSGCUG-NH2, corresponding to the tryptic C-terminal fragment (488-499) of hTrxR. To shed light on the pharmacological differences between AF and AF-AcCys, we carried out a comparative experimental stability study and a theoretical estimation of bond dissociation energies, unveiling the higher strength of the Au-S bond in AF-AcCys. From the results, it emerged that the lower lipophilicity of AF-AcCys with respect to AF could be a key feature for its different antibacterial activity. The differences and similarities between AF and AF-AcCys are discussed, alongside the opportunities and consequences that chemical structure modifications imply.
Collapse
Affiliation(s)
- Lorenzo Chiaverini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via G. Moruzzi, 13, 56124 Pisa, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via G. Moruzzi, 13, 56124 Pisa, Italy
| | - Arianna Nardinocchi
- Department of Biology, University of Pisa, Via San Zeno 35-39, 56100 Pisa, Italy
| | - Iogann Tolbatov
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, 43007 Tarragona, Spain
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università degli Studi "G. D'Annunzio" Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy
| | - Mariagrazia Di Luca
- Department of Biology, University of Pisa, Via San Zeno 35-39, 56100 Pisa, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy
| |
Collapse
|
30
|
Massai L, Cirri D, Marzo T, Messori L. Auranofin and its analogs as prospective agents for the treatment of colorectal cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:1-14. [PMID: 35582525 PMCID: PMC8992591 DOI: 10.20517/cdr.2021.71] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/05/2021] [Accepted: 12/06/2021] [Indexed: 06/06/2023]
Abstract
Today colorectal cancer (CRC) is one of the leading causes of cancer death worldwide. This disease is poorly chemo-sensitive toward the existing medical treatments so that new and more effective therapeutic agents are urgently needed and intensely sought. Platinum drugs, oxaliplatin in particular, were reported to produce some significant benefit in CRC treatment, triggering the general interest of medicinal chemists and oncologists for metal-based compounds as candidate anti-CRC drugs. Within this frame, gold compounds and, specifically, the established antiarthritic drug auranofin with its analogs, form a novel group of promising anticancer agents. Owing to its innovative mechanism of action and its favorable pharmacological profile, auranofin together with its derivatives are proposed here as novel experimental agents for CRC treatment, capable of overcoming resistance to platinum drugs. Some encouraging results in this direction have already been obtained. A few recent studies demonstrate that the action of auranofin may be further potentiated through the preparation of suitable pharmaceutical formulations capable of protecting the gold pharmacophore from unselective reactivity or through the design of highly synergic drug combinations. The perspectives of the research in this field are outlined.
Collapse
Affiliation(s)
- Lara Massai
- Department of Chemistry, University of Florence, Sesto Fiorentino 50019, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa 56124, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
- CISUP - Centre for Instrumentation Sharing (Centro per l’Integrazione della Strumentazione Scientifica), University of Pisa, Pisa 56126, Italy
- University Consortium for Research in the Chemistry of Metal ions in Biological Systems (CIRCMSB), Bari 70126, Italy
| | - Luigi Messori
- Department of Chemistry, University of Florence, Sesto Fiorentino 50019, Italy
| |
Collapse
|
31
|
Ferraro G, Demitri N, Vitale L, Sciortino G, Sanna D, Ugone V, Garribba E, Merlino A. Spectroscopic/Computational Characterization and the X-ray Structure of the Adduct of the V IVO-Picolinato Complex with RNase A. Inorg Chem 2021; 60:19098-19109. [PMID: 34847328 PMCID: PMC8693189 DOI: 10.1021/acs.inorgchem.1c02912] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 12/12/2022]
Abstract
The structure, stability, and enzymatic activity of the adduct formed upon the reaction of the V-picolinato (pic) complex [VIVO(pic)2(H2O)], with an octahedral geometry and the water ligand in cis to the V═O group, with the bovine pancreatic ribonuclease (RNase A) were studied. While electrospray ionization-mass spectrometry, circular dichroism, and ultraviolet-visible absorption spectroscopy substantiate the interaction between the metal moiety and RNase A, electron paramagnetic resonance (EPR) allows us to determine that a carboxylate group, stemming from Asp or Glu residues, and imidazole nitrogen from His residues are involved in the V binding at acidic and physiological pH, respectively. Crystallographic data demonstrate that the VIVO(pic)2 moiety coordinates the side chain of Glu111 of RNase A, by substituting the equatorial water molecule at acidic pH. Computational methods confirm that Glu111 is the most affine residue and interacts favorably with the OC-6-23-Δ enantiomer establishing an extended network of hydrogen bonds and van der Waals stabilizations. By increasing the pH around neutrality, with the deprotonation of histidine side chains, the binding of the V complex to His105 and His119 could occur, with that to His105 which should be preferred when compared to that to the catalytically important His119. The binding of the V compound affects the enzymatic activity of RNase A, but it does not alter its overall structure and stability.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department
of Chemical Sciences, University of Naples
Federico II, I-80126 Napoli, Italy
| | - Nicola Demitri
- Elettra−Sincrotrone
Trieste, S.S. 14 km 163.5
in Area Science Park, 34149 Trieste, Italy
| | - Luigi Vitale
- Department
of Chemical Sciences, University of Naples
Federico II, I-80126 Napoli, Italy
| | - Giuseppe Sciortino
- Institute
of Chemical Research of Catalonia (ICIQ), The Barcelona Institute
of Science and Technology, 43007 Tarragona, Spain
| | - Daniele Sanna
- Istituto
di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Trav. La Crucca 3, I-07100 Sassari, Italy
| | - Valeria Ugone
- Istituto
di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Trav. La Crucca 3, I-07100 Sassari, Italy
| | - Eugenio Garribba
- Dipartimento
di Scienze Mediche, Chirurgiche e Sperimentali, Università di Sassari, Viale San Pietro, I-07100 Sassari, Italy
| | - Antonello Merlino
- Department
of Chemical Sciences, University of Naples
Federico II, I-80126 Napoli, Italy
| |
Collapse
|
32
|
In Vitro Anti-SARS-CoV-2 Activity of Selected Metal Compounds and Potential Molecular Basis for Their Actions Based on Computational Study. Biomolecules 2021; 11:biom11121858. [PMID: 34944502 PMCID: PMC8699537 DOI: 10.3390/biom11121858] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Metal-based drugs represent a rich source of chemical substances of potential interest for the treatment of COVID-19. To this end, we have developed a small but representative panel of nine metal compounds, including both synthesized and commercially available complexes, suitable for medical application and tested them in vitro against the SARS-CoV-2 virus. The screening revealed that three compounds from the panel, i.e., the organogold(III) compound Aubipyc, the ruthenium(III) complex KP1019, and antimony trichloride (SbCl3), are endowed with notable antiviral properties and an acceptable cytotoxicity profile. These initial findings prompted us to perform a computational study to unveil the likely molecular basis of their antiviral actions. Calculations evidenced that the metalation of nucleophile sites in SARS-CoV-2 proteins or nucleobase strands, induced by Aubipyc, SbCl3, and KP1019, is likely to occur. Remarkably, we found that only the deprotonated forms of Cys and Sec residues can react favorably with these metallodrugs. The mechanistic implications of these findings are discussed.
Collapse
|
33
|
Lamarche J, Alcoceba Álvarez E, Cordeau E, Enjalbal C, Massai L, Messori L, Lobinski R, Ronga L. Comparative reactivity of medicinal gold(I) compounds with the cyclic peptide vasopressin and its diselenide analogue. Dalton Trans 2021; 50:17487-17490. [PMID: 34796892 DOI: 10.1039/d1dt03470g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The reactions of the medicinal gold(I) compound auranofin and its close analogues with vasopressin and the diselenide analogue were comparatively investigated by LC-electrospray MS/MS. Evidence is gained of the possible cleavage of the S-S and Se-Se bridges induced by Au(I). Notably, we found that, in the absence of reducing agents, the sulfur and selenium atoms are metallated only at high temperature (70 °C) with the preferential binding of gold to selenium. The reaction with the S-S bridge can take place at physiological temperature (37 °C) under reducing conditions. The implications of these results are discussed in the general frame of the reactivity of biologically relevant soft Lewis acids with peptides and proteins.
Collapse
Affiliation(s)
- Jeremy Lamarche
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Pau, France.
| | | | | | | | - Lara Massai
- Department of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Luigi Messori
- Department of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Ryszard Lobinski
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Pau, France. .,IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Luisa Ronga
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Pau, France.
| |
Collapse
|
34
|
Massai L, Ciambellotti S, Cosottini L, Messori L, Turano P, Pratesi A. Direct detection of iron clusters in L ferritins through ESI-MS experiments. Dalton Trans 2021; 50:16464-16467. [PMID: 34729572 DOI: 10.1039/d1dt03106f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human cytoplasmic ferritins are heteropolymers of H and L subunits containing a catalytic ferroxidase center and a nucleation site for iron biomineralization, respectively. Here, ESI-MS successfully detected labile metal-protein interactions revealing the formation of tetra- and octa-iron clusters bound to L subunits, as previously underscored by X-ray crystallography.
Collapse
Affiliation(s)
- Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Silvia Ciambellotti
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy. .,Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy. .,Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (C.I.R.M.M.P.), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy
| | - Lucrezia Cosottini
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy. .,Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy. .,Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (C.I.R.M.M.P.), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Paola Turano
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy. .,Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy. .,Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (C.I.R.M.M.P.), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy
| |
Collapse
|
35
|
Abdalbari FH, Telleria CM. The gold complex auranofin: new perspectives for cancer therapy. Discov Oncol 2021; 12:42. [PMID: 35201489 PMCID: PMC8777575 DOI: 10.1007/s12672-021-00439-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced stages of cancer are highly associated with short overall survival in patients due to the lack of long-term treatment options following the standard form of care. New options for cancer therapy are needed to improve the survival of cancer patients without disease recurrence. Auranofin is a clinically approved agent against rheumatoid arthritis that is currently enrolled in clinical trials for potential repurposing against cancer. Auranofin mainly targets the anti-oxidative system catalyzed by thioredoxin reductase (TrxR), which protects the cell from oxidative stress and death in the cytoplasm and the mitochondria. TrxR is over-expressed in many cancers as an adaptive mechanism for cancer cell proliferation, rendering it an attractive target for cancer therapy, and auranofin as a potential therapeutic agent for cancer. Inhibiting TrxR dysregulates the intracellular redox state causing increased intracellular reactive oxygen species levels, and stimulates cellular demise. An alternate mechanism of action of auranofin is to mimic proteasomal inhibition by blocking the ubiquitin-proteasome system (UPS), which is critically important in cancer cells to prevent cell death when compared to non-cancer cells, because of its role on cell cycle regulation, protein degradation, gene expression, and DNA repair. This article provides new perspectives on the potential mechanisms used by auranofin alone, in combination with diverse other compounds, or in combination with platinating agents and/or immune checkpoint inhibitors to combat cancer cells, while assessing the feasibility for its repurposing in the clinical setting.
Collapse
Affiliation(s)
- Farah H Abdalbari
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Carlos M Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
36
|
Ok K, Filipovic MR, Michel SLJ. Targeting Zinc Finger Proteins with Exogenous Metals and Molecules: Lessons learned from Tristetraprolin, a CCCH type Zinc Finger. Eur J Inorg Chem 2021; 2021:3795-3805. [PMID: 34867080 PMCID: PMC8635303 DOI: 10.1002/ejic.202100402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Indexed: 11/09/2022]
Abstract
ZF proteins are ubiquitous eukaryotic proteins that play important roles in gene regulation. ZFs contain small domains made up of a combination of four cysteine and histidine residues, and are classified based up on the identity of these residues and their spacing. One emerging class of ZFs are the Cys3His (or CCCH) class of ZFs. These ZFs play key roles in regulating RNA. In this minireview, an overview of the CCCH class of ZFs, with a focus on tristetraprolin (TTP) is provided. TTP regulates inflammation by controlling cytokine mRNAs, and there is an interest in modulating TTP activity to control inflammation. Two methods to control TTP activity are to target with exogenous metals (a 'metals in medicine' approach) or to target with endogenous signaling molecules. Work that has been done to target TTP with Fe, Cu, Cd and Au as well as with H2S is reviewed. This includes attention to new methods that have been developed to monitor metal exchange with the spectroscopically silent ZnII including native electro-spray ionization mass spectrometry (ESI-MS), spin-filter inductively coupled plasma mass spectrometry (ICP-MS) and cryo-electro-spray mass spectrometry (CSI-MS); along with fluorescence anisotropy (FA) to follow RNA binding.
Collapse
Affiliation(s)
- Kiwon Ok
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Milos R Filipovic
- Leibniz-Institut für Analytische, Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
| | - Sarah L J Michel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| |
Collapse
|
37
|
Kou L, Wei S, Kou P. Current Progress and Perspectives on Using Gold Compounds for the Modulation of Tumor Cell Metabolism. Front Chem 2021; 9:733463. [PMID: 34434922 PMCID: PMC8382570 DOI: 10.3389/fchem.2021.733463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/26/2021] [Indexed: 01/14/2023] Open
Abstract
Altered cellular metabolism, which is essential for the growth and survival of tumor cells in a specific microenvironment, is one of the hallmarks of cancer. Among the most significant changes in the metabolic pattern of tumor cells is the shift from oxidative phosphorylation to aerobic glycolysis for glucose utilization. Tumor cells also exhibit changes in patterns of protein and nucleic acid metabolism. Recently, gold compounds have been shown to target several metabolic pathways and a number of metabolites in tumor cells. In this review, we summarize how gold compounds modulate glucose, protein, and nucleic acid metabolism in tumor cells, resulting in anti-tumor effects. We also discuss the rationale underlying the anti-tumor effects of these gold compounds and highlight how to effectively utilize against various types of tumors.
Collapse
Affiliation(s)
- Leiya Kou
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Pei Kou
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, China.,Department of Medical Record, Wuhan No. 1 Hospital, Wuhan, China
| |
Collapse
|
38
|
Grifagni D, Calderone V, Giuntini S, Cantini F, Fragai M, Banci L. SARS-CoV-2 M pro inhibition by a zinc ion: structural features and hints for drug design. Chem Commun (Camb) 2021; 57:7910-7913. [PMID: 34278402 DOI: 10.1039/d1cc02956h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Structural data on the SARS-CoV-2 main protease in complex with a zinc-containing organic inhibitor are already present in the literature and gave hints on the presence of a zinc binding site involving the catalytically relevant cysteine and histidine residues. In this paper, the structural basis of ionic zinc binding to the SARS-CoV-2 main protease has been elucidated by X-ray crystallography. The zinc binding affinity and its ability to inhibit the SARS-CoV-2 main protease have been investigated. These findings provide solid ground for the design of potent and selective metal-conjugated inhibitors of the SARS-CoV-2 main protease.
Collapse
Affiliation(s)
- Deborah Grifagni
- Magnetic Resonance Center (CERM), University of Florence, via Sacconi 6, Sesto Fiorentino, 50019, Italy.
| | - Vito Calderone
- Magnetic Resonance Center (CERM), University of Florence, via Sacconi 6, Sesto Fiorentino, 50019, Italy. and Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, Sesto Fiorentino, 50019, Italy and Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), via Sacconi 6, Sesto Fiorentino, 50019, Italy
| | - Stefano Giuntini
- Magnetic Resonance Center (CERM), University of Florence, via Sacconi 6, Sesto Fiorentino, 50019, Italy.
| | - Francesca Cantini
- Magnetic Resonance Center (CERM), University of Florence, via Sacconi 6, Sesto Fiorentino, 50019, Italy. and Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, Sesto Fiorentino, 50019, Italy and Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), via Sacconi 6, Sesto Fiorentino, 50019, Italy
| | - Marco Fragai
- Magnetic Resonance Center (CERM), University of Florence, via Sacconi 6, Sesto Fiorentino, 50019, Italy. and Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, Sesto Fiorentino, 50019, Italy and Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), via Sacconi 6, Sesto Fiorentino, 50019, Italy
| | - Lucia Banci
- Magnetic Resonance Center (CERM), University of Florence, via Sacconi 6, Sesto Fiorentino, 50019, Italy. and Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3, Sesto Fiorentino, 50019, Italy and Consorzio Interuniversitario Risonanze Magnetiche MetalloProteine (CIRMMP), via Sacconi 6, Sesto Fiorentino, 50019, Italy
| |
Collapse
|
39
|
Fayyaz S, Shaikh M, Gasperini D, Nolan SP, Smith AD, Choudhary MI. In vitro and in cellulo anti-diabetic activity of AuI- and AuIII-isothiourea complexes. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2021.108666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
40
|
Braccini S, Rizzi G, Biancalana L, Pratesi A, Zacchini S, Pampaloni G, Chiellini F, Marchetti F. Anticancer Diiron Vinyliminium Complexes: A Structure-Activity Relationship Study. Pharmaceutics 2021; 13:1158. [PMID: 34452119 PMCID: PMC8398472 DOI: 10.3390/pharmaceutics13081158] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/31/2022] Open
Abstract
A series of 16 novel diiron complexes of general formula [Fe2Cp2(CO)(μ-CO){μ-η1:η3-C(R')C(R″)CN(R)(Y)}]CF3SO3 (2-7), bearing different substituents on the bridging vinyliminium ligand, was synthesized in 69-95% yields from the reactions of diiron μ-aminocarbyne precursors with various alkynes. The products were characterized by elemental analysis, IR, 1H and 13C NMR spectroscopy; moreover the X-ray structures of 2c (R = Y = CH2Ph, R' = R″ = Me) and 3a (R = CH2CH=CH2, Y = R' = Me, R″ = H) were ascertained by single-crystal X-ray diffraction studies. NMR and UV-Vis methods were used to assess the D2O solubility, the stability in aqueous solution at 37 °C and the octanol-water partition coefficients of the complexes. A screening study evidenced a potent cytotoxicity of 2-7 against the A2780 cancer cell line, with a remarkable selectivity compared to the nontumoral Balb/3T3 cell line; complex 4c (R = Cy, Y = R' = R″ = Me) revealed as the most performant of the series. The antiproliferative activity of a selection of complexes was also assessed on the cisplatin-resistant A2780cisR cancer cell line, and these complexes were capable of inducing a significant ROS production. Moreover, ESI-MS experiments indicated the absence of interaction of selected complexes with cytochrome c and the potentiality to inhibit the thioredoxin reductase enzyme (TrxR).
Collapse
Affiliation(s)
- Simona Braccini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Giorgia Rizzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Lorenzo Biancalana
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Stefano Zacchini
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, Viale Risorgimento 4, I-40136 Bologna, Italy;
| | - Guido Pampaloni
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Federica Chiellini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| | - Fabio Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy; (S.B.); (G.R.); (L.B.); (A.P.); (G.P.)
| |
Collapse
|
41
|
Gamberi T, Pratesi A, Messori L, Massai L. Proteomics as a tool to disclose the cellular and molecular mechanisms of selected anticancer gold compounds. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
42
|
Cirri D, Bartoli F, Pratesi A, Baglini E, Barresi E, Marzo T. Strategies for the Improvement of Metal-Based Chemotherapeutic Treatments. Biomedicines 2021; 9:504. [PMID: 34064364 PMCID: PMC8147839 DOI: 10.3390/biomedicines9050504] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
This article provides an overview of the various research approaches we have explored in recent years to improve metal-based agents for cancer or infection treatments. Although cisplatin, carboplatin, and oxaliplatin remain the cornerstones in tumor chemotherapy, the discovery and approval of novel inorganic anticancer drugs is a very slow process. Analogously, although a few promising inorganic drugs have found clinical application against parasitic or bacterial infections, their use remains relatively limited. Moreover, the discovery process is often affected by small therapeutic enhancements that are not attractive for the pharmaceutical industry. However, the availability of increasing mechanistic information for the modes of action of established inorganic drugs is fueling the exploration of various approaches for developing effective inorganic chemotherapy agents. Through a series of examples, some from our own research experience, we focus our attention on a number of promising strategies, including (1) drug repurposing, (2) the simple modification of the chemical structures of approved metal-based drugs, (3) testing novel drug combinations, and (4) newly synthesized complexes coupling different anticancer drugs. Accordingly, we aim to suggest and summarize a series of reliable approaches that are exploitable for the development of improved and innovative treatments.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), Univerisity of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy;
| | - Francesco Bartoli
- Department of Translational Research and of New Surgical and Medical Technologies, Univerisity of Pisa, Via Risorgimento, 36, 56126 Pisa, Italy;
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), Univerisity of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy;
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| |
Collapse
|
43
|
Zoppi C, Massai L, Cirri D, Gabbiani C, Pratesi A, Messori L. Protein metalation by two structurally related gold(I) carbene complexes: An ESI MS study. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2021.120297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
44
|
Menconi A, Marzo T, Massai L, Pratesi A, Severi M, Petroni G, Antonuzzo L, Messori L, Pillozzi S, Cirri D. Anticancer effects against colorectal cancer models of chloro(triethylphosphine)gold(I) encapsulated in PLGA-PEG nanoparticles. Biometals 2021; 34:867-879. [PMID: 33907910 PMCID: PMC8313464 DOI: 10.1007/s10534-021-00313-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 04/21/2021] [Indexed: 12/13/2022]
Abstract
Chloro(triethylphosphine)gold(I), (Et3PAuCl hereafter), is an Auranofin (AF)-related compound showing very similar biological and pharmacological properties. Like AF, Et3PAuCl exhibits potent antiproliferative properties in vitro toward a variety of cancer cell lines and is a promising anticancer drug candidate. We wondered whether Et3PAuCl encapsulation might lead to an improved pharmacological profile also considering the likely reduction of unwanted side-reactions that are responsible for adverse effects and for drug inactivation. Et3PAuCl was encapsulated in biocompatible PLGA–PEG nanoparticles (NPs) and the new formulation evaluated in colorectal HCT-116 cancer cells in comparison to the free gold complex. Notably, encapsulated Et3PAuCl (nano-Et3PAuCl hereafter) mostly retains the cellular properties of the free gold complex and elicits even greater cytotoxic effects in colorectal cancer (CRC) cells, mediated by apoptosis and autophagy. Moreover, a remarkable inhibition of two crucial signaling pathways, i.e. ERK and AKT, by nano-Et3PAuCl, was clearly documented. The implications of these findings are discussed.
Collapse
Affiliation(s)
- Alessio Menconi
- Department of Experimental and Clinical Medicine, University of Florence, Viale G.B. Morgagni 50, 50134, Firenze, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy.
| | - Lara Massai
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via G. Moruzzi 13, 56124, Pisa, Italy
| | - Mirko Severi
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Florence, Viale G.B. Morgagni 50, 50134, Firenze, Italy
| | - Lorenzo Antonuzzo
- Azienda Ospedaliero-Universitaria Careggi, S.C. Oncologia Medica 1, Florence, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Viale G.B. Morgagni 50, 50134, Firenze, Italy. .,DI.V.A.L Toscana S.R.L., Via Madonna del Piano, 6, 50019, Sesto Fiorentino, Italy.
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via G. Moruzzi 13, 56124, Pisa, Italy.
| |
Collapse
|
45
|
Thermodynamic Evaluation of the Interactions between Anticancer Pt(II) Complexes and Model Proteins. Molecules 2021; 26:molecules26082376. [PMID: 33921819 PMCID: PMC8072931 DOI: 10.3390/molecules26082376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/30/2021] [Accepted: 04/15/2021] [Indexed: 12/25/2022] Open
Abstract
In this work, we have analysed the binding of the Pt(II) complexes ([PtCl(4′-phenyl-2,2′:6′,2″-terpyridine)](CF3SO3) (1), [PtI(4′-phenyl-2,2′:6′,2″-terpyridine)](CF3SO3) (2) and [PtCl(1,3-di(2-pyridyl)benzene) (3)] with selected model proteins (hen egg-white lysozyme, HEWL, and ribonuclease A, RNase A). Platinum coordination compounds are intensively studied to develop improved anticancer agents. In this regard, a critical issue is the possible role of Pt-protein interactions in their mechanisms of action. Multiple techniques such as differential scanning calorimetry (DSC), electrospray ionization mass spectrometry (ESI-MS) and UV-Vis absorbance titrations were used to enlighten the details of the binding to the different biosubstrates. On the one hand, it may be concluded that the affinity of 3 for the proteins is low. On the other hand, 1 and 2 strongly bind them, but with major binding mode differences when switching from HEWL to RNase A. Both 1 and 2 bind to HEWL with a non-specific (DSC) and non-covalent (ESI-MS) binding mode, dominated by a 1:1 binding stoichiometry (UV-Vis). ESI-MS data indicate a protein-driven chloride loss that does not convert into a covalent bond, likely due to the unfavourable complexes’ geometries and steric hindrance. This result, together with the significant changes of the absorbance profiles of the complex upon interaction, suggest an electrostatic binding mode supported by some stacking interaction of the aromatic ligand. Very differently, in the case of RNase A, slow formation of covalent adducts occurs (DSC, ESI-MS). The reactivity is higher for the iodo-compound 2, in agreement with iodine lability higher than chlorine.
Collapse
|
46
|
Safir Filho M, Scattolin T, Dao P, Tzouras NV, Benhida R, Saab M, Van Hecke K, Lippmann P, Martin AR, Ott I, Nolan SP. Straightforward synthetic route to gold(i)-thiolato glycoconjugate complexes bearing NHC ligands (NHC = N-heterocyclic carbene) and their promising anticancer activity. NEW J CHEM 2021. [DOI: 10.1039/d1nj02117f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A simple and eco-friendly route to gold–NHC complexes bearing different thiosugars is reported.
Collapse
Affiliation(s)
| | - Thomas Scattolin
- Department of Chemistry and Center for Sustainable Chemistry
- Ghent University
- Krijgslaan 281 (S-3)
- Ghent
- Belgium
| | - Pascal Dao
- Institut de Chimie de Nice
- Université Côte d’Azur
- CNRS
- UMR7272
- Nice
| | - Nikolaos V. Tzouras
- Department of Chemistry and Center for Sustainable Chemistry
- Ghent University
- Krijgslaan 281 (S-3)
- Ghent
- Belgium
| | - Rachid Benhida
- Institut de Chimie de Nice
- Université Côte d’Azur
- CNRS
- UMR7272
- Nice
| | - Marina Saab
- Department of Chemistry and Center for Sustainable Chemistry
- Ghent University
- Krijgslaan 281 (S-3)
- Ghent
- Belgium
| | - Kristof Van Hecke
- Department of Chemistry and Center for Sustainable Chemistry
- Ghent University
- Krijgslaan 281 (S-3)
- Ghent
- Belgium
| | - Petra Lippmann
- Institute of Medicinal and Pharmaceutical Chemistry
- Technische Universität Braunschweig
- Beethovenstr. 55
- Braunschweig
- Germany
| | | | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry
- Technische Universität Braunschweig
- Beethovenstr. 55
- Braunschweig
- Germany
| | - Steven P. Nolan
- Department of Chemistry and Center for Sustainable Chemistry
- Ghent University
- Krijgslaan 281 (S-3)
- Ghent
- Belgium
| |
Collapse
|
47
|
Ghini V, Senzacqua T, Massai L, Gamberi T, Messori L, Turano P. NMR reveals the metabolic changes induced by auranofin in A2780 cancer cells: evidence for glutathione dysregulation. Dalton Trans 2021; 50:6349-6355. [PMID: 33885689 DOI: 10.1039/d1dt00750e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
NMR metabolomics represents a powerful tool to characterize the cellular effects of drugs and gain detailed insight into their mode of action. Here, we have exploited NMR metabolomics to illustrate the changes in the metabolic profile of A2780 ovarian cancer cells elicited by auranofin (AF), a clinically approved gold drug now repurposed as an anticancer agent. An early and large increase in intracellular glutathione is highlighted as the main effect of the treatment accompanied by small but significant changes in the levels of a few additional metabolites; the general implications of these findings are discussed in the frame of the current mechanistic knowledge of AF.
Collapse
Affiliation(s)
- Veronica Ghini
- Center of Magnetic Resonance, University of Florence, via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy. and Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy
| | - Tommaso Senzacqua
- Center of Magnetic Resonance, University of Florence, via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Lara Massai
- Department of Chemistry, University of Florence, via della Lastruccia 3-13, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, viale Morgagni 50, Firenze, Italy
| | - Luigi Messori
- Department of Chemistry, University of Florence, via della Lastruccia 3-13, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Paola Turano
- Center of Magnetic Resonance, University of Florence, via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy. and Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), via Luigi Sacconi 6, 50019, Sesto Fiorentino, Firenze, Italy and Department of Chemistry, University of Florence, via della Lastruccia 3-13, 50019, Sesto Fiorentino, Firenze, Italy.
| |
Collapse
|
48
|
Sciortino G, Maréchal JD, Garribba E. Integrated experimental/computational approaches to characterize the systems formed by vanadium with proteins and enzymes. Inorg Chem Front 2021. [DOI: 10.1039/d0qi01507e] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
An integrated instrumental/computational approach to characterize metallodrug–protein adducts at the molecular level is reviewed. A series of applications are described, focusing on potential vanadium drugs with a generalization to other metals.
Collapse
Affiliation(s)
- Giuseppe Sciortino
- Departament de Química
- Universitat Autònoma de Barcelona
- Cerdanyola del Vallès
- Barcelona 08193
- Spain
| | - Jean-Didier Maréchal
- Departament de Química
- Universitat Autònoma de Barcelona
- Cerdanyola del Vallès
- Barcelona 08193
- Spain
| | - Eugenio Garribba
- Dipartimento di Chimica e Farmacia
- Università di Sassari
- 07100 Sassari
- Italy
| |
Collapse
|
49
|
Ferraro G, Cirri D, Marzo T, Pratesi A, Messori L, Merlino A. The first step of arsenoplatin-1 aggregation in solution unveiled by solving the crystal structure of its protein adduct. Dalton Trans 2020; 50:68-71. [PMID: 33320144 DOI: 10.1039/d0dt04068a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Arsenoplatin-1 (AP-1) is an innovative dual-action anticancer agent that contains a platinum(ii) center coordinated to an arsenous acid moiety. We found that AP-1 spontaneously aggregates in aqueous solutions generating oligomeric species of increasing length. Afterward, we succeeded in solving the crystal structure of the adduct formed between the model protein lysozyme and an early AP-1 oligomer that turned out to be a trimer. Remarkably, this crystal structure traps an early stage of AP-1 aggregation offering detailed insight into the molecular process of the oligomer's growth.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy.
| | | | | | | | | | | |
Collapse
|
50
|
On the Different Mode of Action of Au(I)/Ag(I)-NHC Bis-Anthracenyl Complexes Towards Selected Target Biomolecules. Molecules 2020; 25:molecules25225446. [PMID: 33233711 PMCID: PMC7699860 DOI: 10.3390/molecules25225446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022] Open
Abstract
Gold and silver N-heterocyclic carbenes (NHCs) are emerging for therapeutic applications. Multiple techniques are here used to unveil the mechanistic details of the binding to different biosubstrates of bis(1-(anthracen-9-ylmethyl)-3-ethylimidazol-2-ylidene) silver chloride [Ag(EIA)2]Cl and bis(1-(anthracen-9-ylmethyl)-3-ethylimidazol-2-ylidene) gold chloride [Au(EIA)2]Cl. As the biosubstrates, we tested natural double-stranded DNA, synthetic RNA polynucleotides (single-poly(A), double-poly(A)poly(U) and triple-stranded poly(A)2poly(U)), DNA G-quadruplex structures (G4s), and bovine serum albumin (BSA) protein. Absorbance and fluorescence titrations, mass spectrometry together with melting and viscometry tests show significant differences in the binding features between silver and gold compounds. [Au(EIA)2]Cl covalently binds BSA. It is here evidenced that the selectivity is high: low affinity and external binding for all polynucleotides and G4s are found. Conversely, in the case of [Ag(EIA)2]Cl, the binding to BSA is weak and relies on electrostatic interactions. [Ag(EIA)2]Cl strongly/selectively interacts only with double strands by a mechanism where intercalation plays the major role, but groove binding is also operative. The absence of an interaction with triplexes indicates the major role played by the geometrical constraints to drive the binding mode.
Collapse
|