1
|
Troisi R, Galardo F, Ferraro G, Lucignano R, Picone D, Marano A, Trifuoggi M, Sica F, Merlino A. Cisplatin/Apo-Transferrin Adduct: X-ray Structure and Binding to the Transferrin Receptor 1. Inorg Chem 2024. [PMID: 39711171 DOI: 10.1021/acs.inorgchem.4c04435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Here, we report the X-ray structure of the adduct formed upon reaction of cisplatin, one of the most prescribed anticancer agents for the clinic treatment of solid tumors, with the apo-form of human serum transferrin (hTF). Two Pt binding sites were identified in both molecules of the adduct present in the crystal asymmetric unit: Pt binds close to the side chains of Met256 and Met499 at the N- and C-lobe, respectively. In the crystal structure, the cisplatin moiety bound to Met256 also interacts with Ser616 from a symmetry related molecule. Structural analyses, together with in solution data, demonstrate that the presence of iron does not affect the ability of hTF to bind cisplatin and that the cisplatin binding does not significantly alter the overall conformation of the different forms of the protein that remain able to form a complex with the transferrin receptor 1 (TfR1). These data suggest that the different hTF forms can be used as nanocarriers for targeted (combined) metallodrug delivery.
Collapse
Affiliation(s)
- Romualdo Troisi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Francesco Galardo
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Rosanna Lucignano
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Delia Picone
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Alessandra Marano
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Marco Trifuoggi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Filomena Sica
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, I-80126, Naples, Italy
| |
Collapse
|
2
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
3
|
Xue Q, Yu W, Li JP, He C, Guo Z. Revealing the nature of Pt-based immunotherapy through the lens of neoantigens in cancer. Sci Bull (Beijing) 2024; 69:2314-2318. [PMID: 38670854 DOI: 10.1016/j.scib.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Affiliation(s)
- Qi Xue
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wenhao Yu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jie P Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA; Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
4
|
Wang S, Yuan S, Hu H, Zhang J, Cao K, Wang Y, Liu Y. Reactions of Cisplatin with Thioredoxin-1 Regulate Intracellular Redox Homeostasis. Inorg Chem 2024; 63:11779-11787. [PMID: 38850241 DOI: 10.1021/acs.inorgchem.4c01472] [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: 06/10/2024]
Abstract
Cisplatin is a widely used anticancer drug. In addition to inducing DNA damage, increased levels of reactive oxygen species (ROS) play a significant role in cisplatin-induced cell death. Thioredoxin-1 (Trx1), a redox regulatory protein that can scavenge ROS, has been found to eliminate cisplatin-induced ROS, while elevated Trx1 levels are associated with cisplatin resistance. However, it is unknown whether the effect of Trx1 on the cellular response to cisplatin is due to its direct reaction and how this reaction influences the activity of Trx1. In this work, we performed detailed studies of the reaction between Trx1 and cisplatin. Trx1 is highly reactive to cisplatin, and the catalytic motif of Trx1 (CGPC) is the primary binding site of cisplatin. Trx1 can bind up to 6 platinum moieties, resulting in the structural alteration and oligomerization of Trx1 depending on the degree of platination. Platination of Trx1 inhibits its interaction with ASK1, a Trx1-binding protein that regulates cell apoptosis. Furthermore, the reaction with cisplatin suppresses drug-induced ROS generation, which could be associated with drug resistance. This study provides more insight into the mechanism of action of cisplatin.
Collapse
Affiliation(s)
- Shenghu Wang
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Siming Yuan
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hongze Hu
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jiahai Zhang
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Kaiming Cao
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China
| | - Yu Wang
- School of Chemistry & Chemical Engineering and Environmental Engineering, Weifang University, Weifang 261061, China
| | - Yangzhong Liu
- Key Laboratory of Precision and Intelligent Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
5
|
Jin H, Merz KM. LigandDiff: de Novo Ligand Design for 3D Transition Metal Complexes with Diffusion Models. J Chem Theory Comput 2024; 20:4377-4384. [PMID: 38743854 PMCID: PMC11137811 DOI: 10.1021/acs.jctc.4c00232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Transition metal complexes are a class of compounds with varied and versatile properties, making them of great technological importance. Their applications cover a wide range of fields, either as metallodrugs in medicine or as materials, catalysts, batteries, solar cells, etc. The demand for the novel design of transition metal complexes with new properties remains of great interest. However, the traditional high-throughput screening approach is inherently expensive and laborious since it depends on human expertise. Here, we present LigandDiff, a generative model for the de novo design of novel transition metal complexes. Unlike the existing methods that simply extract and combine ligands with the metal to get new complexes, LigandDiff aims at designing configurationally novel ligands from scratch, which opens new pathways for the discovery of organometallic complexes. Moreover, it overcomes the limitations of current methods, where the diversity of new complexes highly relies on the diversity of available ligands, while LigandDiff can design numerous novel ligands without human intervention. Our results indicate that LigandDiff designs unique and novel ligands under different contexts, and these generated ligands are synthetically accessible. Moreover, LigandDiff shows good transferability by generating successful ligands for any transition metal complex.
Collapse
Affiliation(s)
- Hongni Jin
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Kenneth M. Merz
- Department
of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
6
|
Ferraro G, Lyčková T, Massai L, Štarha P, Messori L, Merlino A. Picoplatin binding to proteins: X-ray structures and mass spectrometry data on the adducts with lysozyme and ribonuclease A. Dalton Trans 2024; 53:8535-8540. [PMID: 38727007 DOI: 10.1039/d4dt00773e] [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: 05/22/2024]
Abstract
The reactivity of the anticancer drug picoplatin (cis-amminedichlorido(2-methylpyridine)platinum(II) complex) with the model proteins hen egg white lysozyme (HEWL) and bovine pancreatic ribonuclease (RNase A) was investigated by electrospray ionisation mass spectrometry (ESI MS) and X-ray crystallography. The data were compared with those previously obtained for the adducts of these proteins with cisplatin, carboplatin and oxaliplatin under the same experimental conditions. ESI-MS data show binding of Pt to both proteins, with fragments retaining the 2-methylpyridine ligand and, possibly, a chloride ion. X-ray crystallography identifies different binding sites on the two proteins, highlighting a different behaviour of picoplatin in the absence or presence of dimethyl sulfoxide (DMSO). Metal-containing fragments bind to HEWL close to the side chains of His15, Asp18, Asp119 and both Lys1 and Glu7, whereas they bind to RNase A on the side chain of His12, Met29, His48, Asp53, Met79, His105 and His119. The data suggest that the presence of DMSO favours the loss of 2-methylpyridine and alters the ability of the Pt compound to bind to the two proteins. With both proteins, picoplatin appears to behave similarly to cisplatin and carboplatin when dissolved in DMSO, whereas it behaves more like oxaliplatin in the absence of the coordinating solvent. This study provides important insights into the pharmacological profile of picoplatin and supports the conclusion that coordinating solvents should not be used to evaluate the biological activities of Pt-based drugs.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso universitario di Monte Sant'Angelo, via Cinthia, 21, 80126, Naples, Italy.
| | - Tereza Lyčková
- Department of Inorganic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 12, 771 46 Olomouc, Czech Republic
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia, 3-13, 50019, Sesto Fiorentino, Florence, Italy
| | - Pavel Štarha
- Department of Inorganic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 12, 771 46 Olomouc, Czech Republic
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia, 3-13, 50019, Sesto Fiorentino, Florence, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso universitario di Monte Sant'Angelo, via Cinthia, 21, 80126, Naples, Italy.
| |
Collapse
|
7
|
Mansour AM, Radacki K, Mostafa GAE, Ali EA, Shehab OR. Antimicrobial properties of triazolato terpyridine Pd(II) and Pt(II) complexes formed by [3+2] cycloaddition coupling reaction. Bioorg Chem 2024; 146:107262. [PMID: 38467092 DOI: 10.1016/j.bioorg.2024.107262] [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: 01/16/2024] [Revised: 02/18/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
Modern classes of antimicrobials are crucial because most drugs in development today are basically antibiotic derivatives. Even though a large number of metal-based compounds have been studied as antimicrobial agents, relatively few studies have examined the antimicrobial properties of Pd(II) and Pt(II) compounds. The [3+2] cycloaddition reactions of [M(N3)L]PF6 (M = Pd(II) and Pt(II); L = 4'-(2-pyridyl)-2,2':6',2″-terpyridine) with 4,4,4-trifluoro-2-butynoic acid ethyl ester gave the corresponding triazolate complexes. The reaction products were fully characterized with a variety of analytical and spectroscopic tools including X-ray crystallographic analysis. The crystal structure of [Pd(triazolatoCF3,COOCH2CH3)L]PF6 provided cut-off evidence that the kinetically formed N1-triazolato isomer favoured the isomerization to the thermodynamically stable N2-analogue. The experimental work was complemented with computational work to get an insight into the nature of the predominant triazolate isomer. The lysozyme binding affinity of the triazolate complexes was examined by mass spectrometry. An analysis of the lysozyme Pd(II) adducts suggests a coordinative covalent mode of binding via the loss of the triazolato ligand. The free ligand and its triazolate complexes displayed selective toxicity against Candida albicans and Cryptococcus neoformans, while no cytotoxicity was observed against the normal human embryonic kidney cell line.
Collapse
Affiliation(s)
- Ahmed M Mansour
- Department of Chemistry, United Arab Emirates University, Al-Ain, United Arab Emirates; Department of Chemistry, Cairo University, Faculty of Science, Gamma Street, Giza, Cairo 12613, Egypt.
| | - Krzysztof Radacki
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Gamal A E Mostafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Essam A Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ola R Shehab
- Department of Chemistry, Cairo University, Faculty of Science, Gamma Street, Giza, Cairo 12613, Egypt
| |
Collapse
|
8
|
Shao X, Xing F, Zhang Y, Lok CN, Che CM. Integrative chemoproteomics reveals anticancer mechanisms of silver(i) targeting the proteasome regulatory complex. Chem Sci 2024; 15:5349-5359. [PMID: 38577372 PMCID: PMC10988589 DOI: 10.1039/d3sc04834a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
Abstract
Silver compounds have favorable properties as promising anticancer drug candidates, such as low side effects, anti-inflammatory properties, and high potential to overcome drug resistance. However, the exact mechanism by which Ag(i) confers anticancer activity remains unclear, which hinders further development of anticancer applications of silver compounds. Here, we combine thermal proteome profiling, cysteine profiling, and ubiquitome profiling to study the molecular mechanisms of silver(i) complexes supported by non-toxic thiourea (TU) ligands. Through the formation of AgTU complexes, TU ligands deliver Ag+ ions to cancer cells and tumour xenografts to elicit inhibitory potency. Our chemical proteomics studies show that AgTU acts on the ubiquitin-proteasome system (UPS) and disrupts protein homeostasis, which has been identified as a main anticancer mechanism. Specifically, Ag+ ions are released from AgTU in the cellular environment, directly target the 19S proteasome regulatory complex, and may oxidize its cysteine residues, thereby inhibiting proteasomal activity and accumulating ubiquitinated proteins. After AgTU treatment, proteasome subunits are massively ubiquitinated and aberrantly aggregated, leading to impaired protein homeostasis and paraptotic death of cancer cells. This work reveals the unique anticancer mechanism of Ag(i) targeting the 19S proteasome regulatory complex and opens up new avenues for optimizing silver-based anticancer efficacy.
Collapse
Affiliation(s)
- Xiaojian Shao
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| | - Fangrong Xing
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| | - Yiwei Zhang
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| | - Chun-Nam Lok
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| | - Chi-Ming Che
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F., Building 17W, Hong Kong Science Park, New Territories Hong Kong P. R. China
| |
Collapse
|
9
|
Merlino A. Metallodrug binding to serum albumin: Lessons from biophysical and structural studies. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
10
|
Arabi A, Cogley MO, Fabrizio D, Stitz S, Howard WA, Wheeler KA. Anticancer Activity of Nonpolar Pt(CH 3) 2I 2{ bipy} is Found to be Superior among Four Similar Organoplatinum(IV) Complexes. J Mol Struct 2023; 1274:134551. [PMID: 36644319 PMCID: PMC9836012 DOI: 10.1016/j.molstruc.2022.134551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The anticancer properties of well-defined molecules serve to bolster the field of metals in medicine. Such compounds, particularly those of platinum and their closely related structural analogs, continue to be potentially highly interesting to researchers and clinicians alike. The four octahedral organoplatinum(IV) compounds [Pt(CH3)2X2{bipy-R 2 }] (X = Br, I; bipy-R 2 = 2,2'-bipyridine, 2,2'-bipyridine-4,4'-dicarboxylic acid) have been isolated and structurally characterized by single-crystal X-ray diffraction. Nuclear magnetic resonance and infrared spectroscopic data are also tabulated as useful reference values. The anticancer potential of each compound was assessed via in vitro MTT assays, using human breast cancer cells (cell line ZR-75-1). EC50 values were determined as 11.5 μM for Pt(CH3)2Br2{bipy}; 3020 μM, for Pt(CH3)2Br2{bipy-(CO 2 H) 2 }; 6.1 μM, for Pt(CH3)2I2{bipy}; and 86.0 μM, for Pt(CH3)2I2{bipy-(CO 2 H) 2 }; for comparison, the EC50 value for cisplatin against the ZR-75-1 cells was 16.4 μM. The most cytotoxic of the four compounds Pt(CH3)2I2{bipy} undergoes reaction with glutathione in a THF/water mixture at 68°C very slowly.
Collapse
Affiliation(s)
- Ameneh Arabi
- Department of Chemistry & Biochemistry, University of Alaska Fairbanks, 900 Yukon Drive, Fairbanks, Alaska 99775, United States
| | - Marcus O. Cogley
- Department of Chemistry & Biochemistry, University of Alaska Fairbanks, 900 Yukon Drive, Fairbanks, Alaska 99775, United States
| | - Daniel Fabrizio
- Department of Chemistry & Biochemistry, University of Alaska Fairbanks, 900 Yukon Drive, Fairbanks, Alaska 99775, United States
| | - Shadrach Stitz
- Department of Chemistry & Biochemistry, University of Alaska Fairbanks, 900 Yukon Drive, Fairbanks, Alaska 99775, United States
| | - William A. Howard
- Department of Chemistry & Biochemistry, University of Alaska Fairbanks, 900 Yukon Drive, Fairbanks, Alaska 99775, United States
| | - Kraig A. Wheeler
- Department of Chemistry, Whitworth University, Spokane, Washington 99251, United States
| |
Collapse
|
11
|
Impact of Hydrophobic Chains in Five-Coordinate Glucoconjugate Pt(II) Anticancer Agents. Int J Mol Sci 2023; 24:ijms24032369. [PMID: 36768690 PMCID: PMC9916762 DOI: 10.3390/ijms24032369] [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] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
This study describes new platinum(II) cationic five-coordinate complexes (1-R,R') of the formula [PtR(NHC)(dmphen)(ethene)]CF3SO3 (dmphen = 2,9-dimethyl-1,10-phenanthroline), containing in their axial positions an alkyl group R (methyl or octyl) and an imidazole-based NHC-carbene ligand with a substituent R' of variable length (methyl or octyl) on one nitrogen atom. The Pt-carbene bond is stable both in DMSO and in aqueous solvents. In DMSO, a gradual substitution of dmphen and ethene is observed, with the formation of a square planar solvated species. Octanol/water partitioning studies have revealed the order of hydrophobicity of the complexes (1-Oct,Me > 1-Oct,Oct > 1-Me,Oct > 1-Me,Me). Their biological activity was investigated against two pairs of cancer and non-cancer cell lines. The tested drugs were internalized in cancer cells and able to activate the apoptotic pathway. The reactivity of 1-Me,Me with DNA and protein model systems was also studied using UV-vis absorption spectroscopy, fluorescence, and X-ray crystallography. The compound binds DNA and interacts in various ways with the model protein lysozyme. Remarkably, structural data revealed that the complex can bind lysozyme via non-covalent interactions, retaining its five-coordinate geometry.
Collapse
|
12
|
Troisi R, Galardo F, Ferraro G, Sica F, Merlino A. Cisplatin Binding to Human Serum Transferrin: A Crystallographic Study. Inorg Chem 2023; 62:675-678. [PMID: 36602395 PMCID: PMC9846693 DOI: 10.1021/acs.inorgchem.2c04206] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The molecular mechanism of how human serum transferrin (hTF) recognizes cisplatin at the atomic level is still unclear. Here, we report the molecular structure of the adduct formed upon the reaction of hTF with cisplatin. Pt binds the side chain of Met256 (at the N-lobe), without altering the protein overall conformation.
Collapse
Affiliation(s)
- Romualdo Troisi
- Department
of Chemical Sciences, University of Naples
Federico II, Complesso Universitario di Monte Sant’Angelo, via Cintia, Naples I-80126, Italy
| | - Francesco Galardo
- Department
of Chemical Sciences, University of Naples
Federico II, Complesso Universitario di Monte Sant’Angelo, via Cintia, Naples I-80126, Italy
| | - Giarita Ferraro
- Department
of Chemical Sciences, University of Naples
Federico II, Complesso Universitario di Monte Sant’Angelo, via Cintia, Naples I-80126, Italy
| | - Filomena Sica
- Department
of Chemical Sciences, University of Naples
Federico II, Complesso Universitario di Monte Sant’Angelo, via Cintia, Naples I-80126, Italy
| | - Antonello Merlino
- Department
of Chemical Sciences, University of Naples
Federico II, Complesso Universitario di Monte Sant’Angelo, via Cintia, Naples I-80126, Italy,
| |
Collapse
|
13
|
Mansouri F, Ortiz D, Dyson PJ. Competitive binding studies of the nucleosomal histone targeting drug, [Ru(η 6-p-cymene)Cl 2(pta)] (RAPTA-C), with oligonucleotide-peptide mixtures. J Inorg Biochem 2023; 238:112043. [PMID: 36370502 DOI: 10.1016/j.jinorgbio.2022.112043] [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: 08/13/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Abstract
Protein crystallography and biochemical assays reveal that the organometallic drug, [Ru(η6-p-cymene)Cl2(pta)] (RAPTA-C), preferentially binds to nucleosomal histone proteins in chromatin. To better understand the binding mechanism we report here a mass spectrometric-based competitive binding study between a model peptide from the acidic patch region of the H2A histone protein (the region where RAPTA-C is known to bind) and an oligonucleotide. In contrast to the protein crystallography and biochemical assays, RAPTA-C preferentially binds to the oligonucleotide, confirming that steric factors, rather than electronic effects, primarily dictate binding of RAPTA-C to histone proteins within the nucleosome.
Collapse
Affiliation(s)
- Farangis Mansouri
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland; Department of Chemistry Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Blvd., Gava Zang, Zanjan 45137-66731, Iran
| | - Daniel Ortiz
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | - Paul J Dyson
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland.
| |
Collapse
|
14
|
Farh MK, Louzi I, Abul-Futouh H, Görls H, Häfner N, Runnebaum IB, Weigand W. Platinum(II) and palladium(II) complexes mediated by β-hydroxy-dithioesters ferrocenyl derivatives: synthesis, characterization and antiproliferative activity. J Sulphur Chem 2022. [DOI: 10.1080/17415993.2022.2152285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Micheal K. Farh
- Department of Inorganic and Analytical Chemistry (IAAC), Friedrich Schiller University Jena, Jena, Germany
- Department of Chemistry, Faculty of Science, Assiut University, Assiut, Egypt
| | - Ikrame Louzi
- Department of Inorganic and Analytical Chemistry (IAAC), Friedrich Schiller University Jena, Jena, Germany
| | - Hassan Abul-Futouh
- Department of Chemistry, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Helmar Görls
- Department of Inorganic and Analytical Chemistry (IAAC), Friedrich Schiller University Jena, Jena, Germany
| | - Norman Häfner
- Department of Gynecology, Jena University Hospital- Friedrich Schiller University Jena, Jena, Germany
| | - Ingo B. Runnebaum
- Department of Gynecology, Jena University Hospital- Friedrich Schiller University Jena, Jena, Germany
| | - Wolfgang Weigand
- Department of Inorganic and Analytical Chemistry (IAAC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
15
|
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
|
16
|
Ferraro G, Paolillo M, Sciortino G, Garribba E, Merlino A. Multiple and Variable Binding of Pharmacologically Active Bis(maltolato)oxidovanadium(IV) to Lysozyme. Inorg Chem 2022; 61:16458-16467. [PMID: 36205235 PMCID: PMC9579999 DOI: 10.1021/acs.inorgchem.2c02690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The interaction with
proteins of metal-based drugs plays a crucial
role in their transport, mechanism, and activity. For an active MLn complex, where L is the organic carrier,
various binding modes (covalent and non-covalent, single or multiple)
may occur and several metal moieties (M, ML, ML2, etc.)
may interact with proteins. In this study, we have evaluated the interaction
of [VIVO(malt)2] (bis(maltolato)oxidovanadium(IV)
or BMOV, where malt = maltolato, i.e., the common name for 3-hydroxy-2-methyl-4H-pyran-4-onato) with the model protein hen egg white lysozyme
(HEWL) by electrospray ionization mass spectrometry, electron paramagnetic
resonance, and X-ray crystallography. The multiple binding of different
V-containing isomers and enantiomers to different sites of HEWL is
observed. The data indicate both non-covalent binding of cis-[VO(malt)2(H2O)] and [VO(malt)(H2O)3]+ and covalent binding of [VO(H2O)3–4]2+ and cis-[VO(malt)2] and other V-containing fragments to the side chains of Glu35,
Asp48, Asn65, Asp87, and Asp119 and to the C-terminal carboxylate.
Our results suggest that the multiple and variable interactions of
potential VIVOL2 drugs with proteins can help
to better understand their solution chemistry and contribute to define
the molecular basis of the mechanism of action of these intriguing
molecules. The interaction of [VIVO(malt)2] (BMOV,
malt = maltolato) with hen egg white lysozyme (HEWL) reveals the multiple
binding of different V-containing isomers and enantiomers to different
sites and non-covalent and covalent binding of cis-[VO(malt)2(H2O)], [VO(malt)(H2O)3]+, [VO(H2O)3−4]2+, and cis-[VO(malt)2] to Glu,
Asp, and Asn residues.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia, I-80126 Napoli, Italy
| | - Maddalena Paolillo
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia, I-80126 Napoli, Italy
| | - Giuseppe Sciortino
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, 43007 Tarragona, Spain
| | - Eugenio Garribba
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, I-07100 Sassari, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia, I-80126 Napoli, Italy
| |
Collapse
|
17
|
Sarkar O, Roy M, Biswal D, Ranjan Pramanik N, Paul S, Drew MGB, Chakrabarti S. Structural Exploration and Protein Binding Efficiencies of Binuclear Dioxidomolybdenum(VI) Complexes Constructed from ONO Chelator and Linear N−N Ditopic Spacer. ChemistrySelect 2022. [DOI: 10.1002/slct.202201412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Oiendrilla Sarkar
- Department of Chemistry University College of Science 92, Acharya Prafulla Chandra Road Kolkata 700009, West Bengal India
| | - Malini Roy
- Department of Chemistry University College of Science 92, Acharya Prafulla Chandra Road Kolkata 700009, West Bengal India
| | - Debanjana Biswal
- Department of Chemistry University College of Science 92, Acharya Prafulla Chandra Road Kolkata 700009, West Bengal India
| | - Nikhil Ranjan Pramanik
- Department of Chemistry Bidhannagar College EB-2 Sector-1, Salt Lake, Kolkata 700064 India
| | - Suvendu Paul
- Department of Chemistry BITS-Pilani Hyderabad Campus Shameerpet Hyderabad 500078, Telangana India
| | - Michael G. B. Drew
- Department of Chemistry The University of Reading Whiteknights Reading RG66AD UK
| | - Syamal Chakrabarti
- Department of Chemistry University College of Science 92, Acharya Prafulla Chandra Road Kolkata 700009, West Bengal India
| |
Collapse
|
18
|
Hernández ÁP, Micaelo A, Piñol R, García-Vaquero ML, Aramayona JJ, Criado JJ, Rodriguez E, Sánchez-Gallego JI, Landeira-Viñuela A, Juanes-Velasco P, Díez P, Góngora R, Jara-Acevedo R, Orfao A, Miana-Mena J, Muñoz MJ, Villanueva S, Millán Á, Fuentes M. Comprehensive and systematic characterization of multi-functionalized cisplatin nano-conjugate: from the chemistry and proteomic biocompatibility to the animal model. J Nanobiotechnology 2022; 20:341. [PMID: 35858906 PMCID: PMC9301860 DOI: 10.1186/s12951-022-01546-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background Nowadays, nanoparticles (NPs) have evolved as multifunctional systems combining different custom anchorages which opens a wide range of applications in biomedical research. Thus, their pharmacological involvements require more comprehensive analysis and novel nanodrugs should be characterized by both chemically and biological point of view. Within the wide variety of biocompatible nanosystems, iron oxide nanoparticles (IONPs) present mostly of the required features which make them suitable for multifunctional NPs with many biopharmaceutical applications. Results Cisplatin-IONPs and different functionalization stages have been broadly evaluated. The potential application of these nanodrugs in onco-therapies has been assessed by studying in vitro biocompatibility (interactions with environment) by proteomics characterization the determination of protein corona in different proximal fluids (human plasma, rabbit plasma and fetal bovine serum),. Moreover, protein labeling and LC–MS/MS analysis provided more than 4000 proteins de novo synthetized as consequence of the nanodrugs presence defending cell signaling in different tumor cell types (data available via ProteomeXchanges with identified PXD026615). Further in vivo studies have provided a more integrative view of the biopharmaceutical perspectives of IONPs. Conclusions Pharmacological proteomic profile different behavior between species and different affinity of protein coating layers (soft and hard corona). Also, intracellular signaling exposed differences between tumor cell lines studied. First approaches in animal model reveal the potential of theses NPs as drug delivery vehicles and confirm cisplatin compounds as strengthened antitumoral agents.
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01546-y.
Collapse
Affiliation(s)
- Ángela-Patricia Hernández
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,Department of Pharmaceutical Sciences. Organic Chemistry Section. Faculty of Pharmacy, University of Salamanca, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Ania Micaelo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Rafael Piñol
- INMA, Institute of Nanoscience and Materials of Aragon, CSIC-University of Zaragoza, 50018, Saragossa, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Marina L García-Vaquero
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - José J Aramayona
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Julio J Criado
- Department of Inorganic Chemistry, Faculty of Chemical Sciences, Plaza de los Caídos S/N, 37008, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Emilio Rodriguez
- Department of Inorganic Chemistry, Faculty of Chemical Sciences, Plaza de los Caídos S/N, 37008, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - José Ignacio Sánchez-Gallego
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Alicia Landeira-Viñuela
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Paula Díez
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Rafael Góngora
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Ricardo Jara-Acevedo
- ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Javier Miana-Mena
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - María Jesús Muñoz
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Sergio Villanueva
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Ángel Millán
- INMA, Institute of Nanoscience and Materials of Aragon, CSIC-University of Zaragoza, 50018, Saragossa, Spain. .,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain.
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain. .,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain. .,Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
19
|
Novel Nickel(II), Palladium(II), and Platinum(II) Complexes with O, S Bidendate Cinnamic Acid Ester Derivatives: An In Vitro Cytotoxic Comparison to Ruthenium(II) and Osmium(II) Analogues. Int J Mol Sci 2022; 23:ijms23126669. [PMID: 35743112 PMCID: PMC9224311 DOI: 10.3390/ijms23126669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 02/04/2023] Open
Abstract
(1) Background: Since the discovery of cisplatin’s cytotoxic properties, platinum(II) compounds have attracted much interest in the field of anticancer drug development. Over the last few years, classical structure−activity relationships (SAR) have been broken by some promising new compounds based on platinum or other metals. We focus on the synthesis and characterization of 17 different complexes with β-hydroxydithiocinnamic acid esters as O,S bidendate ligands for nickel(II), palladium(II), and platinum(II) complexes. (2) Methods: The bidendate compounds were synthesized and characterized using classical methods including NMR spectroscopy, MS spectrometry, elemental analysis, and X-ray crystallography, and their cytotoxic potential was assessed using in vitro cell culture assays. Data were compared with other recently reported platinum(II), ruthenium(II), and osmium(II) complexes based on the same main ligand system. (3) Results: SAR analyses regarding the metal ion (M), and the alkyl-chain position (P) and length (L), revealed the following order of the effect strength for in vitro activity: M > P > L. The highest activities have Pd complexes and ortho-substituted compounds. Specific palladium(II) complexes show lower IC50 values compared to cisplatin, are able to elude cisplatin resistance mechanisms, and show a higher cancer cell specificity. (4) Conclusion: A promising new palladium(II) candidate (Pd3) should be evaluated in further studies using in vivo model systems, and the identified SARs may help to target platinum-resistant tumors.
Collapse
|
20
|
Wang L, Liang YS, Wu ZB, Liu YS, Xiao YH, Hu T, Gao R, Fang J, Liu J, Wu AP. Exploring the interaction between Cry1Ac protein and Zn 2+, Cd 2+ metal ions by fluorescence quenching and molecular docking approaches. CHEMOSPHERE 2022; 297:134105. [PMID: 35245590 DOI: 10.1016/j.chemosphere.2022.134105] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 06/14/2023]
Abstract
Bacillus Thuringiensis (Bt) protein has a strong ability to complex with metal ions, which may increase the transport of metal ions in the soil multi-media system. In this study, the interactions between Cry1Ac protein and metal ions (Zn2+ and Cd2+) were investigated through spectroscopies and molecular docking methods. The spectra results showed that both Zn2+ and Cd2+ quenched the fluorescence intensity of Cry1Ac protein through the static quenching. The binding constants with 4-5 orders of magnitude also indicated the interactions between the ions and the Cry1Ac protein. The thermodynamic analysis showed that hydrogen bonds and van der Waals forces were predominant during the processes. In terms of the Förster non-radiation energy transfer theory, the binding distances between metal ions and Cry1Ac protein were approximately 0.21-0.24 nm, indicating the existence of a non-radiative energy transfer between them. Furthermore, molecular docking revealed that the metal ions participated in ligand binding with the Cry1Ac at the locations Asp569, Thr560, Asn564 and Gln566. The present work provided reasonable models helping us further understand the transport effect of heavy metals in the presence of Cry1Ac. The results could provide mechanistic insights into the nature of metal ions-Cry1Ac interactions and offer important information on the toxicity risk of metal ions-Cry1Ac binding interactions.
Collapse
Affiliation(s)
- Li Wang
- College of Resources and Environment, Hunan Agricultural University and Hunan Provincial Key Laboratory of Rural Ecosystem Health in Dongting Lake Area, Changsha, 410128, PR China
| | - Yun-Shan Liang
- College of Resources and Environment, Hunan Agricultural University and Hunan Provincial Key Laboratory of Rural Ecosystem Health in Dongting Lake Area, Changsha, 410128, PR China.
| | - Zhi-Bin Wu
- College of Resources and Environment, Hunan Agricultural University and Hunan Provincial Key Laboratory of Rural Ecosystem Health in Dongting Lake Area, Changsha, 410128, PR China
| | - Yi-Song Liu
- College of Veterinary Medicine, Hunan Agricultural University and National and Local Union Engineering Research Center of Veterinary Herbal Medicine Resource and Initiative, Changsha, 410128, PR China
| | - Yun-Hua Xiao
- College of Bioscience and Biotechnology, Hunan Agricultural University and Hunan Engineering Laboratory for Pollution Control and Waste Utilization in Swine Production, Changsha, 410128, PR China
| | - Teng Hu
- College of Resources and Environment, Hunan Agricultural University and Hunan Provincial Key Laboratory of Rural Ecosystem Health in Dongting Lake Area, Changsha, 410128, PR China
| | - Rong Gao
- College of Resources and Environment, Hunan Agricultural University and Hunan Provincial Key Laboratory of Rural Ecosystem Health in Dongting Lake Area, Changsha, 410128, PR China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University and Hunan Engineering Laboratory for Pollution Control and Waste Utilization in Swine Production, Changsha, 410128, PR China
| | - Jiao Liu
- College of Resources and Environment, Hunan Agricultural University and Hunan Provincial Key Laboratory of Rural Ecosystem Health in Dongting Lake Area, Changsha, 410128, PR China
| | - Ai Ping Wu
- College of Resources and Environment, Hunan Agricultural University and Hunan Provincial Key Laboratory of Rural Ecosystem Health in Dongting Lake Area, Changsha, 410128, PR China
| |
Collapse
|
21
|
Bai H, Shi J, Guo Q, Wang W, Zhang Z, Li Y, Vennampalli M, Zhao X, Wang H. Spectroscopy, Structure, Biomacromolecular Interactions, and Antiproliferation Activity of a Fe(II) Complex With DPA-Bpy as Pentadentate Ligand. Front Chem 2022; 10:888693. [PMID: 35548676 PMCID: PMC9081768 DOI: 10.3389/fchem.2022.888693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 12/30/2022] Open
Abstract
An Fe(II) complex with DPA-Bpy (DPA-Bpy = N,N-bis(2-pyridinylmethyl)-2,20-bipyridine-6 -methanamine) as the ligand was synthesized and characterized to mimic bleomycin. The binding constants (Kb) of the complex with calf thymus DNA and human serum albumin (HSA) were quantitatively evaluated using fluorescence spectroscopy, with Kb as 5.53×105 and 2.40×104 M−1, respectively; the number of the average binding site (n) is close to 1. The thermodynamic analyses suggested that the electrostatic interactions exist between the complex and DNA, and the hydrogen bonding and Van der Waals force exist for the complex and HSA. The Fe complex exhibits cleavage ability toward pBR322 DNA, and the crystal structure of the HSA Fe complex adduct at 2.4 Å resolution clearly shows that His288 serves as the axial ligand of the Fe center complexed with a pentadentate DPA-Bpy ligand. Furthermore, the cytotoxicity of the complex was evaluated against HeLa cells. Both the Fe complex and HSA Fe complex adduct show obvious effect on cell proliferation with an IC50 of 1.18 and 0.82 μM, respectively; they induced cell apoptosis and arrested cell cycles at S phase. This study provides insight into the plausible mechanism underlying their metabolism and pharmacological activity.
Collapse
Affiliation(s)
- Hehe Bai
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, China
| | - Jia Shi
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, China
| | - Qingyu Guo
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, China
| | - Wenming Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, China
| | - Zhigang Zhang
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, China
| | - Yafeng Li
- The Fifth Hospital (Shanxi Provincial People’s Hospital) of Shanxi Medical University, Taiyuan, China
| | | | - Xuan Zhao
- Department of Chemistry, University of Memphis, Memphis, TN, United States
- *Correspondence: Xuan Zhao, ; Hongfei Wang,
| | - Hongfei Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, China
- *Correspondence: Xuan Zhao, ; Hongfei Wang,
| |
Collapse
|
22
|
Antiproliferative activity and DNA binding studies of cyclometalated complexes of platinum(II) containing 2-vinylpyridine. Biometals 2022; 35:617-627. [PMID: 35445906 DOI: 10.1007/s10534-022-00392-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/04/2022] [Indexed: 11/02/2022]
Abstract
The cytotoxic activity of four cyclometalated platinum(II) complexes [PtMe(vpy)(L)], containing 2-vinylpyridine (vpy) and the phosphine ligands (L) PMe2Ph (1a), PPh3 (1b), PMePh2 (1c), and P(c-Hex)3 (1d), were evaluated against human breast cancer (MDA-MB-231), human lung cancer (A549), human colon cancer (SW1116), and non-tumor epithelial breast (MCF-10 A) cell lines. The highest activity was found for 1c with IC50 values of 21.10 µM, 23.36 µM, and 12.96 µM, compared to cisplatin, which was 10.12 µM, 47.57 µM, and 19.50 µM against the A549, SW1116, and MDA-MB-231 cell lines, respectively. 1a-d showed a higher selectivity index (SI) than cisplatin. Docking studies confirmed interaction to the DNA minor groove for all complexes. Genotoxicity studies on 1c showed interactions with the genomic content of malignant cells. Compared with cisplatin as a positive control, a slight shift was found in the electrophoresis mobility, which was utilized further to study the direct interaction of 1c with DNA.
Collapse
|
23
|
Brink A, Jacobs FJF, Helliwell JR. Trends in coordination of rhenium organometallic complexes in the Protein Data Bank. IUCRJ 2022; 9:180-193. [PMID: 35371500 PMCID: PMC8895017 DOI: 10.1107/s2052252522000665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/18/2022] [Indexed: 06/14/2023]
Abstract
Radiopharmaceutical development has similar overall characteristics to any biomedical drug development requiring a compound's stability, aqueous solubility and selectivity to a specific disease site. However, organometallic complexes containing 188/186Re or 99mTc involve a d-block transition-metal radioactive isotope and therefore bring additional factors such as metal oxidation states, isotope purity and half life into play. This topical review is focused on the development of radiopharmaceuticals containing the radioisotopes of rhenium and technetium and, therefore, on the occurrence of these organometallic complexes in protein structures in the Worldwide Protein Data Bank (wwPDB). The purpose of incorporating the group 7 transition metals of rhenium/technetium in the protein and the reasons for study by protein crystallography are described, as certain PDB studies were not aimed at drug development. Technetium is used as a medical diagnostic agent and involves the 99mTc isotope which decays to release gamma radiation, thereby employed for its use in gamma imaging. Due to the periodic relationship among group 7 transition metals, the coordination chemistry of rhenium is similar (but not identical) to that of technetium. The types of reactions the potential model radiopharmaceutical would prefer to partake in, and by extension knowing which proteins and biomolecules the compound would react with in vivo, are needed. Crystallography studies, both small molecule and macromolecular, are a key aspect in understanding chemical coordination. Analyses of bonding modes, coordination to particular residues and crystallization conditions are presented. In our Forward look as a concluding summary of this topical review, the question we ask is: what is the best way for this field to progress?
Collapse
Affiliation(s)
- Alice Brink
- Chemistry Department, University of the Free State, Nelson Mandela Drive, Bloemfontein, South Africa
- Department of Chemistry, The University of Manchester, Oxford Road, Manchester, United Kingdom
| | - Francois J. F. Jacobs
- Chemistry Department, University of the Free State, Nelson Mandela Drive, Bloemfontein, South Africa
| | - John R. Helliwell
- Department of Chemistry, The University of Manchester, Oxford Road, Manchester, United Kingdom
| |
Collapse
|
24
|
Masaryk L, Zoufalý P, Słoczyńska K, Zahradniková E, Milde D, Koczurkiewicz-Adamczyk P, Štarha P. New Pt(II) diiodido complexes containing bidentate 1,3,4-thiadiazole-based ligands: synthesis, characterization, cytotoxicity. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.120891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
25
|
Tolbatov I, Cirri D, Tarchi M, Marzo T, Coletti C, Marrone A, Messori L, Re N, Massai L. Reactions of Arsenoplatin-1 with Protein Targets: A Combined Experimental and Theoretical Study. Inorg Chem 2022; 61:3240-3248. [PMID: 35137586 PMCID: PMC8864615 DOI: 10.1021/acs.inorgchem.1c03732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Arsenoplatin-1 (AP-1) is a dual-action anticancer metallodrug with a promising pharmacological profile that features the simultaneous presence of a cisplatin-like center and an arsenite center. We investigated its interactions with proteins through a joint experimental and theoretical approach. The reactivity of AP-1 with a variety of proteins, including carbonic anhydrase (CA), superoxide dismutase (SOD), myoglobin (Mb), glyceraldehyde 3-phosphate dehydrogenase (GAPDH), and human serum albumin (HSA), was analyzed by means of electrospray ionization mass spectrometry (ESI MS) measurements. In accordance with previous observations, ESI MS experiments revealed that the obtained metallodrug-protein adducts originated from the binding of the [(AP-1)-Cl]+ fragment to accessible protein residues. Remarkably, in two cases, i.e., Mb and GAPDH, the formation of a bound metallic fragment that lacked the arsenic center was highlighted. The reactions of AP-1 with various nucleophiles side chains of neutral histidine, methionine, cysteine, and selenocysteine, in neutral form as well as cysteine and selenocysteine in anionic form, were subsequently analyzed through a computational approach. We found that the aquation of AP-1 is energetically disfavored, with a reaction free energy of +19.2 kcal/mol demonstrating that AP-1 presumably attacks its biological targets through the exchange of the chloride ligand. The theoretical analysis of thermodynamics and kinetics for the ligand-exchange processes of AP-1 with His, Met, Cys, Sec, Cys-, and Sec- side chain models unveils that only neutral histidine and deprotonated cysteine and selenocysteine are able to effectively replace the chloride ligand in AP-1.
Collapse
Affiliation(s)
- Iogann Tolbatov
- Institut
de Chimie Moleculaire de l’Université de Bourgogne (ICMUB),
Université de Bourgogne Franche-Comté (UBFC), Avenue Alain Savary 9, 21078 Dijon, France
| | - Damiano Cirri
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via G. Moruzzi
13, 56124 Pisa, Italy
| | - Matteo Tarchi
- Department
of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Tiziano Marzo
- Department
of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy
- CISUP
- Centre for Instrumentation Sharing (Centro per l’Integrazione
della Strumentazione Scientifica), University
of Pisa, 56126 Pisa, Italy
- University
Consortium for Research in the Chemistry of Metal ions in Biological
Systems (CIRCMSB), Via
Celso Ulpiani 27, 70126 Bari, Italy
| | - Cecilia Coletti
- Dipartimento
di Farmacia, Università “G
d’Annunzio” di Chieti-Pescara, Via dei Vestini 31, 66013, Chieti, Italy
| | - Alessandro Marrone
- Dipartimento
di Farmacia, Università “G
d’Annunzio” di Chieti-Pescara, Via dei Vestini 31, 66013, Chieti, Italy
| | - Luigi Messori
- Department
of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Nazzareno Re
- Dipartimento
di Farmacia, Università “G
d’Annunzio” di Chieti-Pescara, Via dei Vestini 31, 66013, Chieti, Italy
| | - Lara Massai
- Department
of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
26
|
Relevance of Fluorinated Ligands to the Design of Metallodrugs for Their Potential Use in Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14020402. [PMID: 35214133 PMCID: PMC8874657 DOI: 10.3390/pharmaceutics14020402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/29/2022] Open
Abstract
Fluorination of pharmaceutical agents has afforded crucial modifications to their pharmacological profiles, leading to important advances in medicinal chemistry. On the other hand, metallodrugs are considered to be valuable candidates in the treatment of several diseases, albeit with the caveat that they may exhibit pharmacological disadvantages, such as poor water solubility, low bioavailability and short circulating time. To surmount these limitations, two approaches have been developed: one based on the design of novel metallodrug-delivering carriers and the other based on optimizing the structure of the ligands bound to the metal center. In this context, fluorination of the ligands may bring beneficial changes (physicochemical and biological) that can help to elude the aforementioned drawbacks. Thus, in this review, we discuss the use of fluorinated ligands in the design of metallodrugs that may exhibit potential anticancer activity.
Collapse
|
27
|
Nahaei A, Mandegani Z, Chamyani S, Fereidoonnezhad M, Shahsavari HR, Kuznetsov NY, Nabavizadeh SM. Half-Sandwich Cyclometalated Rh III Complexes Bearing Thiolate Ligands: Biomolecular Interactions and In Vitro and In Vivo Evaluations. Inorg Chem 2022; 61:2039-2056. [PMID: 35023727 DOI: 10.1021/acs.inorgchem.1c03218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A class of cyclometalated RhIII complexes [Cp*Rh(ppy)(SR)] bearing thiolate ligands, Cp* = pentamethylcyclopentadienyl, ppy = 2-phenylpyridinate, and R = pyridyl (Spy, 2), pyrimidyl (SpyN, 3), benzimidazolyl (Sbi, 4), and benzothiazolyl (Sbt, 5), were produced and identified by means of spectroscopic methods. The in vitro cytotoxicity of the RhIII compounds in three different human mortal cancerous cell lines (ovarian, SKOV3; breast, MCF-7; lung, A549) and a normal lung (MRC-5) cell line were evaluated, indicating the selectivity of these cyclometalated RhIII complexes to cancer cells. Complex 5, selected for in vivo experiment, has shown an effective inhibition of tumor growth in SKOV3 xenograft mouse model relative to control (p-values < 0.05 and < 0.01). Importantly, the outcomes of H&E (hematoxylin and eosin) staining and hematological analysis revealed negligible toxicity of 5 compared to cisplatin on a functioning of the main organs of mouse. Molecular docking, UV-vis, and emission spectroscopies (fluorescence, 3D fluorescence, synchronous) techniques were carried out on 1-5 to peruse the mechanism of the anticancer activities of these complexes. The obtained data help to manifest the binding affinity between the rhodium compounds and calf thymus DNA (CT-DNA) through the interaction by DNA minor groove and moderate binding affinity with bovine serum albumin (BSA), particularly with the cavity in the subdomain IIA. It can be concluded that the Rh-thiolate complexes are highly promising leads for the development of novel effective DNA-targeted anticancer drugs.
Collapse
Affiliation(s)
- Asma Nahaei
- Professor Rashidi Laboratory of Organometallic Chemistry, Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71467-13565, Iran
| | - Zeinab Mandegani
- Professor Rashidi Laboratory of Organometallic Chemistry, Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71467-13565, Iran
| | - Samira Chamyani
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Masood Fereidoonnezhad
- Toxicology Research Center; Department of Medicinal Chemistry, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran
| | - Hamid R Shahsavari
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Nikolai Yu Kuznetsov
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilov st. 28, 119991 Moscow, Russian Federation
| | - S Masoud Nabavizadeh
- Professor Rashidi Laboratory of Organometallic Chemistry, Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71467-13565, Iran
| |
Collapse
|
28
|
Simultaneous mass spectrometry analysis of cisplatin with oligonucleotide-peptide mixtures: implications for the mechanism of action. J Biol Inorg Chem 2022; 27:239-248. [PMID: 35064831 PMCID: PMC8907109 DOI: 10.1007/s00775-022-01924-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/30/2021] [Indexed: 11/03/2022]
Abstract
AbstractAlthough genomic DNA is the primary target of anticancer platinum-based drugs, interactions with proteins also play a significant role in their overall activity. In this study, competitive binding of cisplatin with an oligonucleotide and two peptides corresponding to segments of H2A and H2B histone proteins was investigated by mass spectrometry. Following the determination of the cisplatin binding sites on the oligonucleotide and peptides by tandem mass spectrometry, competitive binding was studied and transfer of platinum fragments from the platinated peptides to the oligonucleotide explored. In conjunction with previous studies on the nucleosome, the results suggest that all four of the abundant histone proteins serve as a platinum drug reservoir in the cell nucleus, providing an adduct pool that can be ultimately transferred to the DNA.
Graphical abstract
Collapse
|
29
|
Annunziata A, Ferraro G, Cucciolito ME, Imbimbo P, Tuzi A, Monti DM, Merlino A, Ruffo F. Halo complexes of gold( i) containing glycoconjugate carbene ligands: synthesis, characterization, cytotoxicity and interaction with proteins and DNA model systems. Dalton Trans 2022; 51:10475-10485. [DOI: 10.1039/d2dt00423b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
New neutral Au(i) glycoconjugate carbene complexes show stability in aqueous solutions and interact with both DNA and protein model systems. Cytotoxicity studies demonstrate that the activity depends on the halide ancillary ligand.
Collapse
Affiliation(s)
- Alfonso Annunziata
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia 21, 80126, Napoli, Italy
- Consorzio Interuniversitario di Reattività Chimica e Catalisi (CIRCC), Via Celso Ulpiani 27, 70126, Bari, Italy
| | - Giarita Ferraro
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia 21, 80126, Napoli, Italy
| | - Maria Elena Cucciolito
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia 21, 80126, Napoli, Italy
- Consorzio Interuniversitario di Reattività Chimica e Catalisi (CIRCC), Via Celso Ulpiani 27, 70126, Bari, Italy
| | - Paola Imbimbo
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia 21, 80126, Napoli, Italy
| | - Angela Tuzi
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia 21, 80126, Napoli, Italy
| | - Daria Maria Monti
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia 21, 80126, Napoli, Italy
| | - Antonello Merlino
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia 21, 80126, Napoli, Italy
| | - Francesco Ruffo
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cintia 21, 80126, Napoli, Italy
- Consorzio Interuniversitario di Reattività Chimica e Catalisi (CIRCC), Via Celso Ulpiani 27, 70126, Bari, Italy
| |
Collapse
|
30
|
Mansour AM. Pd(ii) and Pt(ii) complexes of tridentate ligands with selective toxicity against Cryptococcus neoformans and Candida albicans. RSC Adv 2021; 11:39748-39757. [PMID: 35494132 PMCID: PMC9044551 DOI: 10.1039/d1ra06559a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/22/2021] [Indexed: 12/21/2022] Open
Abstract
Novel Pd(ii) and Pt(ii) complexes of the tridentate 2,6-bis(1-ethyl-benzimidazol-2'-yl)pyridine (LBZ), and 4'-(2-pyridyl)-2,2':6',2''-terpyridine (LPY) ligands were synthesized, characterized using a variety of analytical and spectroscopic tools, and screened for their potential antimicrobial properties against some bacterial and fungal strains as well as cytotoxicity against healthy human embryonic kidney (HEK293) cells. The electronic structures of the complexes were investigated by time-dependent density functional theory calculations. The free ligand LPY and benzimidazole complexes exhibited selective toxicity against Cryptococcus neoformans and Candida albicans, while displaying no cytotoxicity against HEK293. In the case of Cryptococcus neoformans, the antifungal activities of the benzimidazole-based complexes (MIC = 1.58-2.62 μM) are higher than those of the reference drug fluconazole (26.1 μM).
Collapse
Affiliation(s)
- Ahmed M Mansour
- Department of Chemistry, Faculty of Science, Cairo University Gamma Street Giza Cairo 12613 Egypt
| |
Collapse
|
31
|
Schijven LMI, Saggiomo V, Velders AH, Bitter JH, Nikiforidis CV. Au 3+-Induced gel network formation of proteins. SOFT MATTER 2021; 17:9682-9688. [PMID: 34633019 DOI: 10.1039/d1sm01031j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The formation of protein gel networks in aqueous systems is a result of protein intermolecular interactions after an energy input, like heating. In this research, we report that a redox reaction between Au3+ ions and proteins can also lead to the formation of a protein gel network. Amino acids, like cysteine and tyrosine, get oxidized and form covalent bonds with neighboring protein molecules, while Au3+ ions get reduced to Au+ and Au0, nucleate and form gold nanoparticles. The protein gel network formation occurs within 2 h at room temperature and can be tuned by varying Au3+/protein ratio and accelerated by increasing the incubation temperature. The proposed Au3+-induced gel network formation was applied to different proteins, like egg yolk high-density lipoprotein, bovine serum albumin and whey protein. This research opens new insights for the investigation of the metal-protein interactions and may aid in the design of novel hybrid-soft nanocomposite materials.
Collapse
Affiliation(s)
- Laura M I Schijven
- Biobased Chemistry and Technology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.
- BioNanoTechnology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Vittorio Saggiomo
- BioNanoTechnology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Aldrik H Velders
- BioNanoTechnology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Johannes H Bitter
- Biobased Chemistry and Technology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.
| | - Constantinos V Nikiforidis
- Biobased Chemistry and Technology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.
| |
Collapse
|
32
|
Löke DR, Helderman RFCPA, Franken NAP, Oei AL, Tanis PJ, Crezee J, Kok HP. Simulating drug penetration during hyperthermic intraperitoneal chemotherapy. Drug Deliv 2021; 28:145-161. [PMID: 33427507 PMCID: PMC7808385 DOI: 10.1080/10717544.2020.1862364] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Hyperthermic intraperitoneal chemotherapy (HIPEC) is administered to treat residual microscopic disease after debulking cytoreductive surgery. During HIPEC, a limited number of catheters are used to administer and drain fluid containing chemotherapy (41–43 °C), yielding heterogeneities in the peritoneum. Large heterogeneities may lead to undertreated areas, increasing the risk of recurrences. Aiming at intra-abdominal homogeneity is therefore essential to fully exploit the potential of HIPEC. More insight is needed into the extent of the heterogeneities during treatments and assess their effects on the efficacy of HIPEC. To that end we developed a computational model containing embedded tumor nodules in an environment mimicking peritoneal conditions. Tumor- and treatment-specific parameters affecting drug delivery like tumor size, tumor shape, velocity, temperature and dose were assessed using three-dimensional computational fluid dynamics (CFD) to demonstrate their effect on the drug distribution and accumulation in nodules. Clonogenic assays performed on RKO colorectal cell lines yielded the temperature-dependent IC50 values of cisplatin (19.5–6.8 micromolar for 37–43 °C), used to compare drug distributions in our computational models. Our models underlined that large nodules are more difficult to treat and that temperature and velocity are the most important factors to control the drug delivery. Moderate flow velocities, between 0.01 and 1 m/s, are optimal for the delivery of cisplatin. Furthermore, higher temperatures and higher doses increased the effective penetration depth with 69% and 54%, respectively. We plan to extend the software developed for this study toward patient-specific treatment planning software, capable of mapping and assist in reducing heterogeneous flow patterns.
Collapse
Affiliation(s)
- Daan R Löke
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Roxan F C P A Helderman
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicolaas A P Franken
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Arlene L Oei
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands
| | - Pieter J Tanis
- Department for Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Johannes Crezee
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H Petra Kok
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Loreto D, Merlino A. The interaction of rhodium compounds with proteins: A structural overview. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213999] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Mansour AM, Radacki K, Shehab OR. Sulfonate improves water solubility and cell selective toxicity and alters the lysozyme binding activity of half sandwich Rh(iii) complexes. Dalton Trans 2021; 50:10701-10706. [PMID: 34337627 DOI: 10.1039/d1dt00979f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Introduction of the propyl-sulfonic acid group at N1 of the coordinated 2-(2-pyridyl)benzimidazole ligand (L) in [RhCl(η5-C5Me5)L](CF3SO3) gives rise to a water-soluble complex, which can bind to the model protein lysozyme via non-covalent interactions. The complex shows selective moderate toxicity against Cryptococcus neoformans (MIC = 21.6-43.3 μM) and exhibits no cytotoxicity to healthy HEK293 cells.
Collapse
Affiliation(s)
- Ahmed M Mansour
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, Giza, Cairo 12613, Egypt.
| | - Krzysztof Radacki
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Ola R Shehab
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, Giza, Cairo 12613, Egypt.
| |
Collapse
|
35
|
Schmidt C, Babu T, Kostrhunova H, Timm A, Basu U, Ott I, Gandin V, Brabec V, Gibson D. Are Pt(IV) Prodrugs That Release Combretastatin A4 True Multi-action Prodrugs? J Med Chem 2021; 64:11364-11378. [PMID: 34342437 DOI: 10.1021/acs.jmedchem.1c00706] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
"Multi-action" Pt(IV) derivatives of cisplatin with combretastatin A4 (CA4) bioactive ligands that are conjugated to Pt(IV) by carbonate are unique because the ligand (IC50 < 10 nM) is dramatically 1000-folds more cytotoxic than cisplatin in vitro. The Pt(IV)-CA4 prodrugs were as cytotoxic as CA4 itself, indicating that the platinum moiety probably plays an insignificant role in triggering cytotoxicity, suggesting that the Pt(IV)-CA4 complexes act as prodrugs for CA4 rather than as true multi-action prodrugs. In vivo tests (Lewis lung carcinoma) show that ctc-[Pt(NH3)2(PhB)(CA4)Cl2] inhibited tumor growth by 93% compared to CA4 (67%), cisplatin (84%), and 1:1:1 cisplatin/CA4/PhB (85%) while displaying <5% body weight loss compared to cisplatin (20%) or CA4 (10%). In this case, and perhaps with other extremely potent bioactive ligands, platinum(IV) acts merely as a self-immolative carrier triggered by reduction in the cancer cell with only a minor contribution to cytotoxicity.
Collapse
Affiliation(s)
- Claudia Schmidt
- Institute for Drug Research, School of Pharmacy, The Hebrew University, 91120 Jerusalem, Israel
| | - Tomer Babu
- Institute for Drug Research, School of Pharmacy, The Hebrew University, 91120 Jerusalem, Israel
| | - Hana Kostrhunova
- Institute of Biophysics, Czech Academy of Sciences, Academy of Sciences, Kralovopolska 135, 61265 Brno, Czech Republic
| | - Annika Timm
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstrasse 55, 38106 Braunschweig, Germany
| | - Uttara Basu
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstrasse 55, 38106 Braunschweig, Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstrasse 55, 38106 Braunschweig, Germany
| | - Valentina Gandin
- Dipartimento di Scienze del Farmaco, Universita di Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Viktor Brabec
- Institute of Biophysics, Czech Academy of Sciences, Academy of Sciences, Kralovopolska 135, 61265 Brno, Czech Republic.,Department of Biophysics, Faculty of Science, Palacky University in Olomouc, Slechtitelu 27, 78371 Olomouc, Czech Republic
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University, 91120 Jerusalem, Israel
| |
Collapse
|
36
|
Ferraro G, Loreto D, Merlino A. Interaction of Platinum-based Drugs with Proteins: An Overview of Representative Crystallographic Studies. Curr Top Med Chem 2021; 21:6-27. [PMID: 32579504 DOI: 10.2174/1568026620666200624162213] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Pt-based drugs are widely used in clinics for the treatment of cancer. The mechanism of action of these molecules relies on their interaction with DNA. However, the recognition of these metal compounds by proteins plays an important role in defining pharmacokinetics, side effects and their overall pharmacological profiles. Single crystal X-ray diffraction studies provided important information on the molecular mechanisms at the basis of this process. Here, the molecular structures of representative adducts obtained upon reaction with proteins of selected Pt-based drugs, including cisplatin, carboplatin and oxaliplatin, are briefly described and comparatively examined. Data indicate that metal ligands play a significant role in driving the reaction of Pt compounds with proteins; non-covalent interactions that occur in the early steps of Pt compound/protein recognition process play a crucial role in defining the structure of the final Pt-protein adduct. In the metallated protein structures, Pt centers coordinate few protein side chains, such as His, Met, Cys, Asp, Glu and Lys residues upon releasing labile ligands.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department of Chemistry Ugo Schiff, University of Florence, Sesto Fiorentino, Firenze, Italy
| | - Domenico Loreto
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy
| |
Collapse
|
37
|
Interference between copper transport systems and platinum drugs. Semin Cancer Biol 2021; 76:173-188. [PMID: 34058339 DOI: 10.1016/j.semcancer.2021.05.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/17/2021] [Indexed: 01/06/2023]
Abstract
Cisplatin, or cis-diamminedichloridoplatinum(II) cis-[PtCl2(NH3)2], is a platinum-based anticancer drug largely used for the treatment of various types of cancers, including testicular, ovarian and colorectal carcinomas, sarcomas, and lymphomas. Together with other platinum-based drugs, cisplatin triggers malignant cell death by binding to nuclear DNA, which appears to be the ultimate target. In addition to passive diffusion across the cell membrane, other transport systems, including endocytosis and some active or facilitated transport mechanisms, are currently proposed to play a pivotal role in the uptake of platinum-based drugs. In this review, an updated view of the current literature regarding the intracellular transport and processing of cisplatin will be presented, with special emphasis on the plasma membrane copper permease CTR1, the Cu-transporting ATPases, ATP7A and ATP7B, located in the trans-Golgi network, and the soluble copper chaperone ATOX1. Their role in eliciting cisplatin efficacy and their exploitation as pharmacological targets will be addressed.
Collapse
|
38
|
Bahkali A, Wei JX, Deng Y. Structural comparison of ethylenediamine platinum(II) complexes containing thiourea and its di- and tetramethyl substituted derivatives. J COORD CHEM 2021. [DOI: 10.1080/00958972.2021.1923015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Ashwaq Bahkali
- Department of Chemistry, Texas Southern University, Houston, TX, USA
| | - Jacob Xin Wei
- Department of Chemistry, Texas Southern University, Houston, TX, USA
| | - Yuanjian Deng
- Department of Chemistry, Texas Southern University, Houston, TX, USA
| |
Collapse
|
39
|
Godzieba M, Ciesielski S. Natural DNA Intercalators as Promising Therapeutics for Cancer and Infectious Diseases. Curr Cancer Drug Targets 2021; 20:19-32. [PMID: 31589125 DOI: 10.2174/1568009619666191007112516] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/11/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023]
Abstract
Cancer and infectious diseases are one of the greatest challenges of modern medicine. An unhealthy lifestyle, the improper use of drugs, or their abuse are conducive to the increase of morbidity and mortality caused by these diseases. The imperfections of drugs currently used in therapy for these diseases and the increasing problem of drug resistance have forced a search for new substances with therapeutic potential. Throughout history, plants, animals, fungi and microorganisms have been rich sources of biologically active compounds. Even today, despite the development of chemistry and the introduction of many synthetic chemotherapeutics, a substantial part of the new compounds being tested for treatment are still of natural origin. Natural compounds exhibit a great diversity of chemical structures, and thus possess diverse mechanisms of action and molecular targets. Nucleic acids seem to be a good molecular target for substances with anticancer potential in particular, but they may also be a target for antimicrobial compounds. There are many types of interactions of small-molecule ligands with DNA. This publication focuses on the intercalation process. Intercalators are compounds that usually have planar aromatic moieties and can insert themselves between adjacent base pairs in the DNA helix. These types of interactions change the structure of DNA, leading to various types of disorders in the functioning of cells and the cell cycle. This article presents the most promising intercalators of natural origin, which have aroused interest in recent years due to their therapeutic potential.
Collapse
Affiliation(s)
- Martyna Godzieba
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Sloneczna 45 G, 10-917 Olsztyn, Poland
| | - Slawomir Ciesielski
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Sloneczna 45 G, 10-917 Olsztyn, Poland
| |
Collapse
|
40
|
Synthesis and characterization of ethylenediamine platinum(II) complexes containing thiourea derivatives. X-ray crystal structures of [Pt(en)(2-imidazolidinethione)2](NO3)2 and [Pt(en)(1-phenyl-2-thiourea)2](NO3)2. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2021.120302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
41
|
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
|
42
|
Ultrasmall iron oxide nanoparticles cisplatin (IV) prodrug nanoconjugate: ICP-MS based strategies to evaluate the formation and drug delivery capabilities in single cells. Anal Chim Acta 2021; 1159:338356. [PMID: 33867043 DOI: 10.1016/j.aca.2021.338356] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/21/2022]
Abstract
Ultrasmall iron oxide nanoparticles (<10 nm) were explored here as nanotransporters of cis-diamminetetrachloroplatinum (IV) (a cisplatin prodrug) in cellular models. The coating of the particles containing reactive carboxylic acid groups enabled the formation of a stable conjugate between the prodrug and the nanoparticles using one pot reaction. The nanoconjugate was characterized by different techniques exhibiting diameters of about 6.6 ± 1.0 nm. The use of a hyphenated strategy based on high performance liquid chromatography (HPLC) coupled to inductively coupled plasma mass spectrometry (ICP-MS) permitted the quantitative evaluation of Fe and Pt in the nanoconjugate. Furthermore, the cellular uptake of the synthetic nanoconjugate was explored by single cell-ICP-MS (SC-ICP) which was used for the first time in this type of studies. The experiments in A2780 and A2780cis, sensitive and resistant ovarian cancer cell models respectively, revealed intracellular platinum concentrations of 12 fg/cell and 4 fg/cell, respectively which were 4-fold higher with respect to the uptake of cisplatin in both models. Intracellular drug release from the nanoconjugate was proved by measuring DNA platination in the same cells. In this case, levels of about 250 ng Pt/mg DNA were observed, about 5-fold higher when the nanoconjugate was used in comparison to cisplatin. Furthermore, the differences between the two lines turned to be significantly smaller than in the case of using cisplatin. The quantitative analytical tools developed here provided essential information required to fully characterize the developed nanoplatforms particularly important to overcome drug resistance.
Collapse
|
43
|
Pereira SAP, Bobbink FD, Dyson PJ, Saraiva MLMFS. Automatic evaluation of cyclooxygenase 2 inhibition induced by metal-based anticancer compounds. J Inorg Biochem 2021; 218:111399. [PMID: 33706122 DOI: 10.1016/j.jinorgbio.2021.111399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/27/2022]
Abstract
An automatic methodology based on micro sequential injection analysis coupled to a lab-on-valve system (termed μSIA-LOV) was developed and used to determine the ability of metal-based anticancer compounds to inhibit cyclooxygenase 2 (COX-2) activity. COX-2 may be involved in pathogenesis of cancer and it is overexpressed in several types of solid tumors. Since platinum-based compounds are extensively used in the treatment of cancer, and ruthenium compounds are considered as promising candidates for next generation of non-targeted anticancer drugs, it is interesting to establish whether COX-2 inhibition is relevant to their mode of action. The μSIA-LOV system was optimized and the IC50 values of each compound were calculated. All the results present RSD values less than 2.5%. IC50 values of 9.7 ± 0.6 μM to 207 ± 3 μM were obtained, with the most active inhibitor for COX-2 being rofecoxib with the metal compounds exhibiting IC50 values in the range 13.7 ± 1.6 to 207 ± 3. The results obtained in this work provide significant information about the mechanism of the studied compounds, mostly ruthenium-based compounds, and the role of COX-2 in their mode of action. Moreover, this work confirmed the potential of the μSIA-LOV system as a simple, versatile, robust, and rapid analytical tool for automating the determination of IC50 values of metal-based compounds.
Collapse
Affiliation(s)
- Sarah A P Pereira
- REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Felix D Bobbink
- Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - M Lúcia M F S Saraiva
- REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| |
Collapse
|
44
|
The Protein-Binding Behavior of Platinum Anticancer Drugs in Blood Revealed by Mass Spectrometry. Pharmaceuticals (Basel) 2021; 14:ph14020104. [PMID: 33572935 PMCID: PMC7911130 DOI: 10.3390/ph14020104] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin and its analogues are widely used as chemotherapeutic agents in clinical practice. After being intravenously administrated, a substantial amount of platinum will bind with proteins in the blood. This binding is vital for the transport, distribution, and metabolism of drugs; however, toxicity can also occur from the irreversible binding between biologically active proteins and platinum drugs. Therefore, it is very important to study the protein-binding behavior of platinum drugs in blood. This review summarizes mass spectrometry-based strategies to identify and quantitate the proteins binding with platinum anticancer drugs in blood, such as offline high-performance liquid chromatography/inductively coupled plasma mass spectrometry (HPLC–ICP-MS) combined with electrospray ionization mass spectrometry (ESI-MS/MS) and multidimensional LC–ESI-MS/MS. The identification of in vivo targets in blood cannot be accomplished without first studying the protein-binding behavior of platinum drugs in vitro; therefore, relevant studies are also summarized. This knowledge will further our understanding of the pharmacokinetics and toxicity of platinum anticancer drugs, and it will be beneficial for the rational design of metal-based anticancer drugs.
Collapse
|
45
|
Wang Z, Fang L, Zhao J, Gou S. Insight into the antitumor actions of sterically hindered platinum(ii) complexes by a combination of STD NMR and LCMS techniques. Metallomics 2021; 12:427-434. [PMID: 32022072 DOI: 10.1039/c9mt00258h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Sterically hindered platinum(ii) complexes have shown great advantages in overcoming platinum drug resistance. In this study, the antitumor actions of sterically hindered platinum(ii) complex 1 (cis-dichloro[(1R,2R)-N1-(2-fluorobenzyl)-1,2-diaminocyclohexane-N,N']platinum(ii), C13H19FPtCl2) were investigated by using saturation transfer difference nuclear magnetic resonance (STD NMR) and liquid chromatography-mass spectrometry (LCMS) techniques. STD NMR was applied to study the HSA (human serum albumin) binding properties, while the interactions between guanosine 5'-monophosphate (5'-GMP) and complex 1 were studied by LCMS. For HSA binding experiments, strong STD signals were observed for protons of sterically hindered parts of carrier ligands, indicating that the sterically hindered moieties of the carrier ligand could be situated inside the binding pocket of HSA. A 19F NMR experiment indicated that complex 1 could interact with HSA. Furthermore, the binding modes of complex 1 with guanosine 5'-monophosphate (5'-GMP) were studied in the absence and presence of glutathione by LCMS. According to the HPLC profiles, a mono-functional binding mode was observed for complex 1 both in the presence and in the absence of glutathione, while a bi-adduct was observed for Pt(DACH)Cl2, which may be one of the reasons for their different biological activities. Hence, this study demonstrated that the NMR method combined with the LCMS technique could provide valuable information to understand the transport and the underlying anticancer mechanisms of the platinum(ii) complex at the molecular level. Moreover, the results reported here can help to reveal the binding mechanisms of the sterically hindered platinum(ii) compounds with biomolecules, which may shed light on the design of novel platinum(ii) anticancer agents with suitable sterically hindered groups.
Collapse
Affiliation(s)
- Zhimei Wang
- Research Center and School of Chemistry and Chemical Engineering, and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China.
| | - Lei Fang
- Research Center and School of Chemistry and Chemical Engineering, and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China.
| | - Jian Zhao
- Research Center and School of Chemistry and Chemical Engineering, and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China.
| | - Shaohua Gou
- Research Center and School of Chemistry and Chemical Engineering, and Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China.
| |
Collapse
|
46
|
Adams M, Sullivan MP, Tong KKH, Goldstone DC, Hanif M, Jamieson SMF, Hartinger CG. Mustards-Derived Terpyridine-Platinum Complexes as Anticancer Agents: DNA Alkylation vs Coordination. Inorg Chem 2021; 60:2414-2424. [PMID: 33497565 DOI: 10.1021/acs.inorgchem.0c03317] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The development of bifunctional platinum complexes with the ability to interact with DNA via different binding modes is of interest in anticancer metallodrug research. Therefore, we report platinum(II) terpyridine complexes to target DNA by coordination and/or through a tethered alkylating moiety. The platinum complexes were evaluated for their in vitro antiproliferative properties against the human cancer cell lines HCT116 (colorectal), SW480 (colon), NCI-H460 (non-small cell lung), and SiHa (cervix) and generally exhibited potent antiproliferative activity although lower than their respective terpyridine ligands. 1H NMR spectroscopy and/or ESI-MS studies on the aqueous stability and reactivity with various small biomolecules, acting as protein and DNA model compounds, were used to establish potential modes of action for these complexes. These investigations indicated rapid binding of complex PtL3 to the biomolecules through coordination to the Pt center, while PtL4 in addition alkylated 9-ethylguanine. PtL3 was investigated for its reactivity to the model protein hen egg white lysozyme (HEWL) by protein crystallography which allowed identification of the Nδ1 atom of His15 as the binding site.
Collapse
Affiliation(s)
- Muneebah Adams
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Matthew P Sullivan
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Kelvin K H Tong
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - David C Goldstone
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Muhammad Hanif
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
47
|
Abstract
Recent advances in structural studies unveiling the basis of the metal compounds/protein recognition process are discussed.
Collapse
Affiliation(s)
- Antonello Merlino
- Department of Chemical Sciences
- University of Naples Federico II
- Complesso Universitario di Monte Sant’Angelo
- Napoli
- Italy
| |
Collapse
|
48
|
Ventura G, Calvano CD, Losito I, Viola A, Cinquepalmi V, Cataldi TRI. In vitro reactions of a cyanocobalamin-cisplatin conjugate with nucleoside monophosphates. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34:e8945. [PMID: 32910479 DOI: 10.1002/rcm.8945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/13/2020] [Accepted: 09/04/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE Cisplatin (CP) is a widely used anticancer drug characterized by toxic side effects that could be alleviated using novel delivery systems including CP prodrugs. The in vitro incubation of a putative prodrug, obtained from cyanocobalamin (CNCbl) and cis-diamminemonochloroplatinum(II) (mCP), with nucleoside monophosphates (NMPs) was investigated. METHODS The in vitro reactions between the putative prodrug CNCbl-mCP and the NMPs of adenosine (AMP), guanosine (GMP), cytidine (CMP) and uridine (UMP) were carried out in slightly acidic water-methanol solutions at 37°C for 24 h. Each sample was examined using reversed-phase liquid chromatography coupled with electrospray ionization in positive ion mode and tandem mass spectrometry (RPLC/ESI-MS/MS) by collision-induced dissociation in a linear ion-trap mass spectrometer. RESULTS Seven adducts were recognized as formed by substitution reactions of the chloride ligand in planar CP. Comparison between observed and theoretical isotopic patterns together with MS/MS fragmentation pathways revealed the presence of single or multiple binding sites depending on the NMP involved. The CNCbl-mCP conjugate was found to interact with N7 or O4 atoms of GMP and UMP, respectively, generating single adducts, while two isomeric adducts were observed for CMP. Finally, AMP gave rise to three isomeric adducts. CONCLUSIONS In agreement with literature data relevant to the interaction between CP and NMPs, the most reactive nucleotides were AMP and GMP. The present RPLC/ESI-MS/MS approach is very promising for investigation of the reactions of CP conjugates with ribonucleotides not only in vitro but also in vivo.
Collapse
Affiliation(s)
- Giovanni Ventura
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, Italy
| | - Cosima Damiana Calvano
- Centro Interdipartimentale SMART, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, Italy
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, Italy
| | - Ilario Losito
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, Italy
- Centro Interdipartimentale SMART, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, Italy
| | - Andrea Viola
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, Italy
| | - Valeria Cinquepalmi
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, Italy
| | - Tommaso R I Cataldi
- Dipartimento di Chimica, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, Italy
- Centro Interdipartimentale SMART, Università degli Studi di Bari Aldo Moro, via Orabona 4, Bari, 70126, 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
|
Daubit IM, Sullivan MP, John M, Goldstone DC, Hartinger CG, Metzler-Nolte N. A Combined Spectroscopic and Protein Crystallography Study Reveals Protein Interactions of Rh I(NHC) Complexes at the Molecular Level. Inorg Chem 2020; 59:17191-17199. [PMID: 33180473 DOI: 10.1021/acs.inorgchem.0c02438] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
While most Rh-N-heterocyclic carbene (NHC) complexes currently investigated in anticancer research contain a Rh(III) metal center, an increasing amount of research is focusing on the cytotoxic activity and mode of action of square-planar [RhCl(COD)(NHC)] (where COD = 1,5-cyclooctadiene) which contains a Rh(I) center. The enzyme thioredoxin reductase (TrxR) and the protein albumin have been proposed as potential targets, but the molecular processes taking place upon protein interaction remain elusive. Herein, we report the preparation of peptide-conjugated and its nonconjugated parent [RhCl(COD)(NHC)] complexes, an in-depth investigation of both their stability in solution, and a crystallographic study of protein interaction. The organorhodium compounds showed a rapid loss of the COD ligand and slow loss of the NHC ligand in aqueous solution. These ligand exchange reactions were reflected in studies on the interaction with hen egg white lysozyme (HEWL) as a model protein in single-crystal X-ray crystallographic investigations. Upon treatment of HEWL with an amino acid functionalized [RhCl(COD)(NHC)] complex, two distinct rhodium adducts were found initially after 7 d of incubation at His15 and after 4 weeks also at Lys33. In both cases, the COD and chlorido ligands had been substituted with aqua and/or hydroxido ligands. While the histidine (His) adduct also indicated a loss of the NHC ligand, the lysine (Lys) adduct retained the NHC core derived from the amino acid l-histidine. In either case, an octahedral coordination environment of the metal center indicates oxidation to Rh(III). This investigation gives the first insight on the interaction of Rh(I)(NHC) complexes and proteins at the molecular level.
Collapse
Affiliation(s)
- Isabelle M Daubit
- Inorganic Chemistry I - Bioinorganic Chemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, Universitätsstr. 150, 44801 Bochum, Germany
| | - Matthew P Sullivan
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Milena John
- Inorganic Chemistry I - Bioinorganic Chemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, Universitätsstr. 150, 44801 Bochum, Germany
| | - David C Goldstone
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Nils Metzler-Nolte
- Inorganic Chemistry I - Bioinorganic Chemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, Universitätsstr. 150, 44801 Bochum, Germany
| |
Collapse
|