Next Article in Journal
Artificial Intelligence and Machine Learning Technology Driven Modern Drug Discovery and Development
Next Article in Special Issue
Semi-Targeted Profiling of Bile Acids by High-Resolution Mass Spectrometry in a Rat Model of Drug-Induced Liver Injury
Previous Article in Journal
Design, Synthesis and Pharmacological Evaluation of New Quinoline-Based Panx-1 Channel Blockers
Previous Article in Special Issue
Recent Advances in Health Benefits of Bioactive Compounds from Food Wastes and By-Products: Biochemical Aspects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Biological Activity of Novel Organotin Compounds with a Schiff Base Containing an Antioxidant Fragment

by
Taisiya A. Antonenko
1,
Yulia A. Gracheva
1,
Dmitry B. Shpakovsky
1,
Mstislav A. Vorobyev
2,
Dmitrii M. Mazur
1,
Victor A. Tafeenko
1,
Yury F. Oprunenko
1,
Elena F. Shevtsova
3,
Pavel N. Shevtsov
3,
Alexey A. Nazarov
1,* and
Elena R. Milaeva
1
1
Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
2
Institute of Geography of the Russian Academy of Sciences, Department of Glaciology, 117312 Moscow, Russia
3
Institute of Physiologically Active Compounds of Russian Academy of Sciences, 142432 Chernogolovka, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2024; https://doi.org/10.3390/ijms24032024
Submission received: 28 December 2022 / Revised: 16 January 2023 / Accepted: 17 January 2023 / Published: 19 January 2023
(This article belongs to the Special Issue 23rd Anniversary of IJMS: Advances in Biochemistry)

Abstract

:
A series of novel organotin(IV) complexes on the base of 2-(N-3′,5′-di-tert-butyl-4′-hydroxyphenyl)-iminomethylphenol (L) of formulae Me2SnBr2(L)2 (1), Bu2SnCl2(L)2(2), Ph2SnCl2(L) (3), Ph2SnCl2(L)2 (4) Ph3SnBr(L)2 (5) were synthesized and characterized by 1H, 13C, 119Sn NMR, IR, ESI-MS and elemental analysis. The crystal structures of initial L and complex 2 were determined by XRD method. It was found that L crystallizes in the orthorhombic syngony. The distorted octahedron geometry around Sn center is observed in the structure of complex 2. Intra- and inter-molecular hydrogen bonds were found in both structures. The antioxidant activity of new complexes as reducing agents, radical scavengers and lipoxygenase inhibitors was estimated spectrophotometrically in CUPRAC and DPPH tests (compounds 1 and 5 were found to be the most active in both methods), and in the process of enzymatic oxidation in vitro of linoleic acid under the action of lipoxygenase LOX 1-B (EC50 > 33.3 μM for complex 2). Furthermore, compounds 1–5 have been investigated for their antiproliferative activity in vitro towards HCT-116, MCF-7 and A-549 and non-malignant WI-38 human cell lines. Complexes 2 and 5 demonstrated the highest activity. The plausible mechanisms of the antiproliferative activity of compounds, including the influence on the polymerization of Tb+MAP, are discussed. Some of the synthesized compounds have also actively induced apoptosis and blocked proliferation in the cell cycle G2/M phase.

1. Introduction

It is known that platinum compounds were the first drugs based on metal compounds that were widely used in cancer treatment therapy. Nevertheless, they are highly toxic and show poor selectivity for different cell lines, and many types of cancer have developed resistance to them. Therefore, the search for new organometallic compounds with anti-tumor activity is an urgent task [1,2,3]. A significant part of this large class of compounds is represented by organotin complexes. It is known that tin complexes with coordination number >4 have a wide spectrum of biological activity, such as fungicidal, antibacterial, nematicidal, insecticidal, herbicidal, anti-inflammatory and anti-tumor activities [4]. It was found that the structure of anti-tumor drugs based on tin compounds is characterized by the presence of a stable ligand-metal bond resistant to hydrolysis. Higher cytotoxicity is manifested when the coordination environment of the tin atom is completely filled (CN = 6) and when the Sn–N and Sn–S bonds are the shortest [5].
Schiff bases common organic ligands. They can be applied as dyes, catalysts, intermediates, polymer stabilizers and pigments. These compounds also demonstrate various types of biological activities, e.g., antimalarial, anti-inflammatory, antiviral and antiproliferative ones [6]. Complexes of Fe(III), Co(II), Zn(II), and Zr(IV) with Schiff base gemifloxacin exhibited promising antifungal activity [7]. Also, there is a study reporting significant biological activity of enrofloxacin Schiff base complexes (H2Erx-en) of Fe(III), Y(III), Zr(IV) and La(III) that demonstrated extremely significant antibacterial activity data [8]. The Cu (II) complex with Gat-o-phdn Schiff base (4E,4′E)-4,4′-(1,2-phenylenebis (azaneylylidene))bis(1-cyclopropyl-6-fluoro-8-methoxy-7-(3-methylpiperazin-1-yl)-1,4-dihydroquinoline-3-carboxylic acid demonstrated very high antimicrobial activity against Staphylococcus aureus [9].
One of the proposed mechanisms of organotins’ action may be due to Sn atom’s ability to bind to sulfhydryl groups, followed by DNA destructionthat ultimately leads to cell death [10]. The toxicity of organotins can also induce oxidative stress in living organisms. To decrease their unwanted high toxicity, it is proposed to use antioxidants; 2,6-dialkylphenols are known to be vitamin E mimetics and inhibitors of radical oxidative processes [11,12]. We found previously that organotin complexes containing Schiff base with 2,6-dialkylphenol fragment demonstrate both good antioxidant and cytotoxic activities [13].
The purpose of this work was the synthesis of novel organotin complexes 15 with Schiff base L containing an antioxidant fragment of 2,6-di-tert-butylphenol (Scheme 1) and the study of their biological activity. Herein we report the antioxidant and antiproliferative properties of new complexes 15.

2. Results and Discussion

2.1. Synthesis and Characterization

Novel complexes 14 were obtained by the interaction of L with organotin chlorides Bu2SnCl2, Ph2SnCl2 and bromide Me2SnBr2 (Scheme 1) in methylene chloride with stirring and low heating (40 °C). The new complex 5 (Scheme 1) was synthesized by the interaction of L with Ph3SnBr in a mixture of chloroform and ethanol. Compounds 15 were found to be stable in air and were explored by elemental analysis, IR, 1H, 13C and 119Sn NMR spectroscopy.
The IR, 1H and 13C spectroscopic data (Figures S1–S10) are comparable with those for structurally similar complexes [13].The IR spectra of compounds 15 demonstrate absorption bands in the region of 3492–3643 cm−1, appropriating to the stretching vibrations of the O-H bond of the sterically hindered non-associated phenolic group.
The proton signal of the phenol group is absent in the 1H NMR spectra of complexes 15, whereas for the starting compound L the signal is observed at 13.64 ppm due to solvent exchange. The spin-spin interaction of H-Sn (Ph2Sn fragment,2JSn-H = 80 Hz) is also found in the 1H NMR spectrum of complex 4.
The chemical shifts in 119Sn spectra varied in the wide interval (from −326 to +112 ppm) depending on the environment of Sn atom (Figures S11–S15).
Compounds 15 were also characterized by HRMS. It turns out that during electrospray ionization, the complex structure is broken, and the charge stays on the ligand part only. There is however indirect evidence, which is described below, in favor of the proposed structure coming from HRMS data. It appears that some intermolecular reactions or redox reactions besides just ionization processes may take place during ESI. The ligand environment, hence, could be significantly changed through the loss of some ligands or by substitution reactions. Compound 1 forms ions m/z 326.2101 C21H28O2N, m/z 474.1431 C23H32O2NSn (Me2SnL), m/z 799.3463 C44H59O4N2Sn (Me2SnL2) (Figure 1). Base peak m/z 326.2104 (C21H28O2N) corresponds to the protonated molecule of ligand.
Similar ions are observed in the mass spectrum of compound 2 with m/z 326.2101 C21H28O2N (L), m/z 558.2370 C29H44O2NSn (Bu2SnL), m/z 594.2129 C29H45O2NClSn (Bu2SnClL), m/z 883.4399 C50H71O4N2Sn (Bu2SnL2) (Figure S16).
In the case of compound 3 two clusters of ions are observed at m/z 594–602 and m/z 672–680, which the isotopic distribution and accurate masses of which nicely fit the formulae.C33H36O2NSn (Ph2SnL) and C39H42O2NSn (Ph3SnL) correspondingly, result from loss of chloride ligands. Signals of low abundance within m/z 612–636 were assigned for C33H37O2NClSn (Ph2SnClL) and C33H38O3NSn (Ph2SnOHL) (Figure S17).
The base peak in the mass spectrum of compound 4 is similar tothat for 3 since they both have the same ligand (Figure S18). However, together with the ions mentioned above there are signals in the m/z 800–950 range (e.g., m/z 923.3742 corresponding to Ph2SnL2, i.e., C54H63O4N2Sn) bringing evidence to the bis-ligand nature of the compound 4. Substitution of Cl-ligands for Br doesn’t significantly change the mass spectrum of compound 5 (Figure S19). The most abundant peaks are the same. The bromine atom is missing in observed signals, but all the rest of the moieties are revealed: m/z 598.1732 C33H36O2NSn (Ph2SnL), m/z 676.2203 C39H42O2NSn (Ph3SnL), m/z 854.4114 C48H66O4N2Sn, m/z 923.3744 C54H63O4N2Sn (Ph2SnL2). Though the studied complexes did not form any molecular species ([M+H]+, [M+Na]+, etc.) under ESI conditions, the ions described above still are useful and reveal the proposed molecular structure.

2.2. Crystal Structures

Recrystallization of compound L from a mixture of petroleum ether and CH2Cl2 gave orange crystals, which were used for crystallographic analysis (Table 1).
It has been established that the ligand crystallizes in the orthorhombic syngony. The N1-C7 bond distance is 1.272(3) Å. X-ray diffraction analysis (XRD) showed the presence of an intramolecular hydrogen bond O2-H32N1 (H32N1 = 1.47 Å, O2-H32N1= 2.54 Å, angle O2-H32-N1 = 150.95°), as well as an intermolecular hydrogen bond O1-H31O2 (H31O2 = 2.13 Å, O1-H31O2 = 2.80 Å, angle O1-H31-O2 = 137.23°).
The angle between the ring plane of the 2,6-di-tert-butylphenol fragment (C1C2C3C4C5C6) and the ring plane of the unhindered phenol group (C8C9C10C11C12C13) is 22.26°. The molecular structure of the compound is shown in Figure 2. Table 2 demonstrates the individual bond distances and angles of L.
As a result of the recrystallization of compound 2 from hexane, orange single crystals, suitable for XRD, were obtained. The coordination number around the tin atom in crystals (L)2Bu2SnCl2 (2) is 6; the coordination polyhedron is a distorted octahedron. The butyl groups, chlorine atoms, and oxygen atoms of the unhindered phenolic group are in a trans-position with respect to each other in the inner coordination sphere of the tin atom (Figure 3a).
Due to proton transfer from the oxygen atom O2 to the nitrogen atom N1, intramolecular hydrogen bonds N1–H21⋯O2 are formed in the structure (H21⋯O2 = 1.80 Å, N1–H21⋯O2 = 2.58 Å, angle N1–H21-O2 = 137.85°) (Figure 3b). It should be noted that, in comparison with the initial ligand, in the structure of the obtained complex, the angle between the ring plane of the 2,6-di-tert-butylphenol fragment and the ring plane of the unhindered phenol group in the structure of 2 decreases significantly and amounts to 4.50°, i.e., these fragments lie almost in the same plane. The individual bond distances and angles of 2 are listed in Table 2.

2.3. CUPRAC Assay (Cu2+ Reducing)

One of the methods for studying the antioxidant activity of compounds is to research their ability to one-electron reduction using the spectrophotometric CUPRAC test. Neocuproine (2,9-dimethyl-1,10-phenanthroline) forms a complex with Cu+ in the presence of antioxidants with an absorption maximum at 450 nm [14]. The experiment was carried out to increase the optical density of a solution of the complex in ethanol. Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid) was used as a standard. The results are presented as TEAC values (Trolox equivalents antioxidant capacity, Table 3). TEAC for Trolox is 1.00 ± 0.03. It was shown that among the most active complexes are compounds 1, 2, 4 and 5.

2.4. DPPH Radical Scavenging Activity

A method for studying the antioxidant (radical scavenging) activity of compounds is their ability to reduce the stable 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical by hydrogen atom transfer [15].
The activity of tin compounds 15 in the DPPH test, as well as the initial ligand L, was studied spectrophotometrically by measuring the decrease in the optical density of DPPH at a wavelength of 517 nm for 1 h. The value of the EC50 parameter (the effective concentration of the compound required to reduce the concentration of the DPPH radical by 50%) was determined graphically according to the dependence of the content of the remaining DPPH (in %) on the primary concentration of compounds (0.01–0.1 mM). EC50 values are presented in Table 4.
Since compounds 15 showed rather high activity, their reaction kinetics were studied, which corresponds to the second order equation. The rate constants k (for each concentration) are obtained from the plot of 1/[DPPH] vs. time (Table 4). It is known that k for L at a concentration of 0.06 mM is 10.9 L mol−1 s−1 [13]. At concentrations higher than 0.04 mM the rate of the DPPH reduction was too high to be evaluated.

2.5. Lipoxygenase Inhibition Assessment

Lipoxygenase LOX 1-B is a plant enzyme that belongs to the class of iron-containing oxygenases and catalyzes the stereospecific oxidation of polyunsaturated fatty acids in the cell, including linoleic acid. It is known that lipoxygenase is able to participate in the destruction of biomembranes [16]. Therefore, the ability of a compound to inhibit this enzyme may indicate its potential antioxidant and anti-inflammatory properties. The antioxidant properties of the obtained compounds were also evaluated in the process of enzymatic oxidation of linoleic acid under the action of lipoxygenase LOX 1-B in vitro. The experiment was carried out by spectrophotometry measuring the content of the oxidation product of linoleic acid—the corresponding hydroperoxides at λmax 234 nm. It was found that the majority of the compounds promote the oxidation of linoleic acid, while complex 2 is only a moderate inhibitor of lipoxygenase (EC50 > 33.3 μM). The results are listed in Table 5.

2.6. Antiproliferative Activity

The antiproliferative activity of compounds 15 was also studied. The compounds were investigated against HCT-116 (human colon cancer), MCF-7 (human breast cancer), A-549 (adenocarcinoma human alveolar basal epithelial) and WI-38 (diploid human cell line composed of fibroblasts) cells using the MTT test [17]. The IC50 values were determined in comparison with cisplatin (Table 6).
It is known that, depending on the organic substituent nature, the cytotoxicity of organotin compounds RnSnX4-nL decreases in accordance with n-Bu > Ph, Et > Me [18,19].
Among the obtained tin complexes, the triphenyltin complex 5 exhibits nanomolar activity (Table 6) that is consistent with the results of our previous works concerning triphenyltin complexes based on 2,6-di-tert-butyl-4-mercaptophenol which exhibited the high activity related with the high lipophilicity of Ph3Sn fragment. In addition, complexes based on dibutyltin and diphenyltin 24 also demonstrate high antiproliferative activity (Figure 4). The activity of obtained complexes against nonmalignant cells WI-38 is comparable to that of HCT-116 cancer cells.

2.7. Cell Death and Cell Cycle Analysis

Apoptosis, known as programmed cell death, is a carefully controlled, energy-dependent process. Cells initiate intracellular processes by responding to specific induction signals. This leads to characteristic physiological changes, which include the externalization of phosphatidylserine (PS) to the cell surface. PS is a membrane component that is usually localized on the inside of the cell membrane. At the beginning of the apoptotic pathway, PS molecules move to the outer surface of the cell membrane, where they can easily be bound by annexin V, a calcium-dependent phospholipid-binding protein with a high affinity for PS. To prove the hypothesis that antiproliferative activity correlates with the induction of apoptosis by organotin compounds, we studied the apoptotic profile of selected cancer cells. The mechanism of cell death was monitored as Annexin V/7-AAD reactivity for compounds 2 and 5 in HCT-116 cells using the Muse®AnnexinV& Dead Cell Kit (Luminex Corp., Austin, TX, USA) by flow cytometry [20]. Cells were incubated with compounds and cisplatin for 24 and 48 h at concentrations corresponding to 2·IC50 values. Both compounds were shown to induce apoptosis actively. The overall percentage of apoptotic cells is 42.6 and 26.8% for 2 and 5, respectively. Interestingly, for compound 2 the main fraction of cells was found in late apoptosis (32.4%). The percentage of apoptotic cells did not exceed 10.3% in the control group (Figure 5). Thereby, HCT-116 cells turned out to be more sensitive to the action of compound 2 after 24 h of incubation.
An overall increase in apoptotic cells for studied compounds was observed after 48 h, e.g., for 2 their number approaches 65%. The fraction of cells in late apoptosis was 46.3% for 2 (Figure 6).
The cell cyclecan be considered the smallest part of the life cycle. It is the series of steps of growth and development that a cell goes through between its birth and reproduction—dividing to form two new daughter cells. For cell division, several tasks must be taken: the cell must grow (G1 phase), copy its genetic material (S phase), prepare to divide (G2 phase), and divide (by mitosis, or M phase).
The assay uses propidium iodide (PI) based staining of DNA content to discriminate and measure the percentage of cells in each cell cycle phase (G0/G1, S, and G2/M). PI discriminates cells at different stages of the cell cycle, based on differential DNA content in the presence of RNAse to increase the specificity of DNA staining.
The effect of compounds 2 and 5 on cell cycle arrest by flow cytometry was carried out. HCT-116 cells were treated for 24 h with compounds at the concentrations of IC50. It was found that compounds block proliferation in the G2/M phase of the cell cycle, preparation for mitosis-mitosis (Figure 7).

2.8. Tubulin Polymerization

It is known that microtubules and their main component tubulin are the prospectivetargets of antiproliferative chemotherapy drugs. In our previous works [11,21] we demonstrated that cytotoxic organotin complexes containing 2,6-di-tert-butylphenol moieties are able to interact with tubulin SH groups. These results suggest that the antiproliferative anti-tumor effects of these compounds may be associated with the disruption of microtubule assembly due to the inhibition of tubulin polymerization in the presence of organotin complexes.
In this work, we studied the influence of the most active compounds 2 and 5 on the crude preparation of Tb+MAPpolymerization. It was found that both compounds affect the guanosine triphosphate (GTP) dependent polymerization of Tb+MAP; some stimulation of polymerization is observed for compound 5 and for compound 2 there is a significant blockade of this process that indicates a potential antiproliferative activity (Figure 8). Both stimulation and blockade of the process of tubulin polymerization can be mechanisms of the antiproliferative activity of the compounds, disrupting the dynamic processes of the cell division microtubules spindle [22].

3. Materials and Methods

3.1. Reagents and Materials

All solvents used were of reagent grade and starting organotin compounds Bu2SnCl2, Ph2SnCl2, Me2SnBr2,Ph3SnBr (Sigma-Aldrich, Merck KGaA, Darmstadt, Germany, 98%), Trolox (Acros Organics, Geel, Belgium, 97%) were used as supplied. DPPH, neocuproine (2,9-dimethyl-1,10-phenanthroline, 99%) xanthine, EDTA, nitroblue tetrazolium (NBT), xanthine oxidase (0.04 MU) and bovine serum albumin were bought from Sigma-Aldrich.
Infrared absorption spectra were registered using KBr pellets on IR200 (Thermo Nicolet Corporation, Madison, WI, USA) spectrophotometer with Fourier transform. The NMR spectra were obtained using a Bruker Avance-400 (Brucker, Karlsruhe, Germany) spectrometer at different frequencies: 400.1 MHz (1H), 100.6 MHz (13C) and 149.15 MHz (119Sn) in CDCl3and DMSO-d6. Orbitrap Elite (Thermo Fisher Scientific, Waltham, MA, USA) mass-spectrometer equipped with an electrospray ionization source (ESI) was used for all high-resolution mass-spectrometry (HRMS) experiments. All compounds dissolved in methanol were directly introduced into the ionization source by means of a syringe pump at 5 μL/min. ESI mass spectra were recorded using positive ionization mode with spray voltage set to 3.4 kV. Xcalibur software (Thermo Xcalibur 3.0.63, Thermo Scientific, Waltham, MA, USA)was used as a system control tool as well as for data collection and data processing. Vaporizer and ion transfer tube temperatures were set to 40 °C and 275 °C correspondingly. All spectra were recorded during 30 s in the m/z range 150–1500. Elemental analyses were carried out using a Vario Micro Cube analyzer (Elementar, Berlin, Germany). The determination of the antioxidant activity of the compounds was investigated on an Evolution 300 UV-Visible (Thermo Scientific, Waltham, MA, USA) cuvette spectrophotometer and a microplate (96 wells) spectrophotometer Multiskan Go (Thermo Fisher Sci., Waltham, MA, USA). The MTT test was carried out on a Zenyth200rt (Anthos, Biochrom, Cambridge, UK) multiwall-plate reader.

3.2. Synthesis

Schiff base 2-(-N-3,5-di-tert-butyl-4-hydroxyphenyl)iminomethylphenol (L), was obtained according to the well-known method [23]. Synthesis of complexes 15 is demonstrated in Scheme 1.

3.2.1. Synthesis and Characterization of Me2SnBr2(L)2 (1)

A solution of 25 mg (0.07 mmol) Me2SnBr2 in 2 mL of CH2Cl2was added to a solution of 50.9 mg (0.148 mmol) of L in 3 mL of CH2Cl2. Then the mixture was stirred at 40 °C for 4 h. The precipitate formed was separated and rinsed with petroleum ether followed by air drying for 24 h. Yield 75 mg (73%);orange-yellow powder; m.p. 135–137 °C. Anal. Calcd for C44H60N2O4Br2Sn (959.47) (%): C, 55.08; H, 6.30; N, 2.92. Found (%): C, 55.32; H, 6.43; N, 2.78. IR(KBr, ν, cm−1): ν(OH) 3627 (m); ν(C-H) 2873–2962 (s); ν(C=N) 1632 (m); 1608; 1486; 1432; 1240; 1157; 766. 1HNMR (CDCl3, 400.1 MHz, δ, ppm): 1.38 (s, 6H, Sn(CH3)2, 2JSn-H = 64 Hz); 1.49 (s, 36H, 4C(CH3)3); 5.29 (s, 2H, 2OH); 6.91 (dd, 2H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 7.01 (d, 2H, Ar, 3JH-H = 8 Hz); 7.16 (s, 4H, 2C6H2); 7.34–7.41 (m, 4H, Ar), 8.60 (s, 2H, 2CH=N). 13CNMR (CDCl3, 100.6 MHz, δ, ppm): 7.52 (Sn-CH3); 29.80 (C(CH3)3); 34.13 (C(CH3)3); 116.86; 117.48; 118.40; 118.93; 131.55; 132.28; 136.58; 138.71; 146.66; 152.85 (CAr); 159.34 (CH=N). 119SnNMR (DMSO-d6, 149.15 MHz, δ, ppm): −326.37.

3.2.2. Synthesis and Characterization of Bu2SnCl2(L)2 (2)

25 mg (0.07 mmol) of Bu2SnCl2 was added to a solution of 48 mg (0.16 mmol) of L in 7 mL of CH2Cl2. The mixture was stirred at 40 °C for 1.5 h. The solvent was moved away in a vacuum. The resulting oil was treated with hexane. The precipitate formed was rinsed with hexane followed by air drying for 24 h. To obtain crystals suitable for XRD, the substance was recrystallized from hexane. Yield 120 mg (85%);bright orange crystals; m.p. 118–120 °C. Anal. Calcd for C50H72N2O4Cl2Sn (954.73) (%): C, 62.90; H, 7.60; N, 2.93. Found (%): C, 62.74; H, 7.52; N, 2.65. IR (KBr, ν, cm−1): ν(OH) 3492 (s); ν(C-H) 2866–2992 (s); ν(C=N) 1641 (s); 1607; 1487; 1432; 1224; 1105; 756. 1H NMR (CDCl3, 400.1 MHz, δ, ppm): 0.94 (t, 6H, 2CH2(CH2)2CH3, 3JH-H = 8 Hz); 1.40–1.46 (m, 4H, 2CH2(CH2)2CH3); 1.49 (s, 36H, 4C(CH3)3); 1.77–1.84 (m, 8H, 2CH2(CH2)2CH3); 5.30 (s, 2H, 2OH); 6.92 (dd, 2H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 7.01 (d, 2H, Ar, 3JH-H = 8 Hz); 7.16 (s, 4H, 2C6H2); 7.34 (dd, 2H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 7.39 (d, 2H, Ar, 3JH-H = 8 Hz); 8.60 (s, 2H, 2CH=N). 13C NMR (CDCl3, 100.6 MHz, δ, ppm): 13.09 (CH3CH2); 25.89 (CH2); 26.29 (CH2); 26.47 (CH2); 29.79 (C(CH3)3); 34.12 (C(CH3)3); 116.79; 117.49; 118.42; 119; 131.47; 132.15; 136.55; 139.54; 152.81; 159.38 (CAr); 160.81 (CH=N). 119Sn NMR (CDCl3, 149.15 MHz, δ, ppm): 112.52.

3.2.3. Synthesis and Characterization of Ph2SnCl2(L) (3)

126 mg (0.39 mmol) of L was added to a solution of 133 mg (0.39 mmol) of Ph2SnCl2 in 8 mL of CH2Cl2. The mixture was stirred at 40 °C for 2 h. The solvent was moved away in a vacuum. The precipitate formed was rinsed with hexane followed by air drying for 24 h.
Yield 258 mg (88%); colouryellow-orange powder;m.p. 76–78°C. Anal. Calcd for C33H37NO2Cl2Sn (669.26) (%): C, 59.22; H, 5.57; N, 2.09. Found (%): C, 59.03; H, 5.35; N, 1.85. Selected IR data (KBr, ν, cm−1): ν(OH) 3615 (s); ν(C-H) 2873–3052 (s); ν(C=N) 1633 (s); 1608; 1484; 1431; 1226; 1151; 731. 1H NMR (CDCl3, 400.1 MHz, δ, ppm): 1.49 (s, 18H, 2C(CH3)3); 5.33 (s, 1H, OH); 6.88 (dd, 1H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 6.97 (d, 1H, Ar, 3JH-H = 8 Hz); 7.20 (s, 4H, 2C6H2); 7.33 (dd, 1H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 7.38 (d, 1H, Ar, 3JH-H = 8 Hz); 7.52–7.55 (m, 6H, 2Sn-Ph); 7.72–7.75 (m, 4H, 2Sn-Ph, 2JSn-H = 80 Hz), 8.57 (s, 1H, 2CH=N). 13C NMR (CDCl3, 100.6 MHz, δ, ppm): 29.79 (C(CH3)3); 34.15 (C(CH3)3); 117.41; 118.21; 122.51; 128.13; 129.21; 131.25; 131.77; 132.84; 134.71; 136.70; 137.17; 139.81; 153.02 (CAr); 159.06 (CH=N). 119Sn NMR (DMSO-d6, 149.15 MHz, δ, ppm): −267.40.

3.2.4. Synthesis and Characterization of Ph2SnCl2(L)2 (4)

107 mg (0.33 mmol) of L and 4 mLof CH2Cl2 were added to a solution of 57 mg (0.165 mmol) of Ph2SnCl2 in 4 mLof CH2Cl2. The mixture was stirred at 40 °C for 3 h. The solvent was moved away in a vacuum. The precipitate formed was rinsed with hexane followed by air-drying for 24 h.
Yield 164 mg (94%); orange powder; m.p.87–89°C. Anal. Calcd for C54H64N2O4Cl2Sn (994.71) (%): C, 65.20; H, 6.49; N, 2.82. Found (%): C, 64.97; H, 6.34; N, 2.76. IR (KBr, ν, cm−1): ν(OH) 3617 (s); ν(C-H) 2872–3053 (s); ν(C=N) 1634 (s); 1612; 1486; 1432; 1228; 1152; 759. 1H NMR (CDCl3, 400.1 MHz, δ, ppm): 1.49 (s, 36H, 4C(CH3)3); 5.31 (s, 2H, 2OH); 6.90 (dd, 2H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 7.00 (d 2H, Ar, 3JH-H = 8 Hz); 7.17 (s, 4H, 2C6H2); 7.33 (dd, 2H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 7.38 (d, 2H, Ar, 3JH-H = 8 Hz); 7.52–7.55 (m, 6H, 2Sn-Ph); 7.72–7.75 (m, 4H, 2Sn-Ph), 8.57 (s, 2H, 2CH=N). 13C NMR (CDCl3, 100.6 MHz, δ, ppm): 29.72 (C(CH3)3); 34.08 (C(CH3)3); 116.51; 117.32; 118.39; 118.95; 126.01; 129.29; 131.41; 131.93; 134.82; 136.96; 139.49; 152.80; 159.29 (CAr); 160.49 (CH=N). 119Sn NMR (DMSO-d6, 149.15 MHz, δ, ppm): −237.08.

3.2.5. Synthesis and Characterization of Ph3SnBr(L)2 (5)

64 mg (0.15 mmol) of Ph3SnBr was added to a solution of 50 mg (0.155 mmol) of L in 4 mL of CHCl3. The mixture was stirred at 40 °C for 1 h. Then it was filtered through a paper filter. The solvent from the filtrate was moved away in a vacuum. The precipitate formed was rinsed with hexane followed by air drying for 24 h.
Yield 85 mg (88%); yellow powder; m.p.99–101°C. Anal. Calcd for C60H69N2O4BrSn (1080.82) (%): C, 66.68; H, 6.43; N, 2.59. Found (%): C, 66.39; H, 6.31; N, 2.42. IR (KBr, ν, cm−1): ν(OH) 3616 (w); ν(C-H) 2872–3067 (s); ν(C=N) 1635 (w); 1613; 1481; 1431; 1234; 1153; 731. 1H NMR (CDCl3, 400.1 MHz, δ, ppm): 1.49 (s, 36H, 4C(CH3)3); 5.29 (s, 2H, 2OH); 6.92 (dd, 2H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 7.01 (d, 2H, Ar, 3JH-H = 8 Hz); 7.16 (s, 4H, 2C6H2); 7.35 (dd, 2H, Ar, 3JH-H = 8 Hz, 3JH-H = 8 Hz); 7.35–7.41 (m, 4H, 4Ar); 7.46–7.49 (m, 9H, 3Sn-Ph); 7.67–7.70 (m, 6H, 3Sn-Ph), 8.60 (s, 2H, 2CH=N). 13C NMR (CDCl3, 100.6 MHz, δ, ppm): 29.79 (C(CH3)3); 34.12 (C(CH3)3); 116.79; 117.50; 118.44; 119.03; 120.68; 128.75; 130.08; 131.45; 132.11; 135.73; 136.41; 136.55; 152.8; 159.41(CAr); 160.73 (CH=N). 119Sn NMR (DMSO-d6, 149.15 MHz, δ, ppm): −262.64.

3.3. Crystallographic Data Collection and Structure Determination

All diffraction data were collected on a STOE StadiVari Pilatus 100 K diffractometer (Stoe&Cie, Darmstadt, Germany) [λ(MoKα) = 0.71073 Å, λ(CuKα) = 1.5418 Å, ω-scans] at 293 K [24] The primary processing of the experimental data array was performed using the WinGX program package [25]. The structures were solved by direct methods and refined by full-matrix least-squares procedures on F2 using SHELXL97 [26]. All non-hydrogen atoms were refined anisotropically, and hydrogen atoms were located at calculated positions and refined via the ‘riding model’. Crystal data and structure refinement parameters are listed in Table 1. CCDC2205597 (L), 2205598 (2) contain the supplementary crystallographic data for this paper. The structures of complexes were drawn using the MERCURY CSD 3.1 program [27].

3.4. CUPRAC Assay (Cu2+ Reducing)

The method proposed by Apak et al. was used with slight modification [14]. For this reason, 0.05 mLof CuCl2 solution (0.01 M), 0.05 mLof methanol neocuproine solution (7.5 mM) and 0.25 mLof ammonium acetate buffer solution (1 M) were added to a test tube, followed by mixing with different concentrations of tested compounds (10–100 μM). The mixtures were kept at room temperature. Absorbance was measured at 450 nm on Multiskan Go microplate spectrophotometer (Thermo Fisher Sci., Waltham, MA, USA) against a reagent blank 35 min later. The increase of reaction mixture absorbance in comparison with control indicates the reduction capability of the test compound. Results were presented in Trolox-equivalents. TEAC CUPRAC values were obtained graphically using absorbance data and a linear calibration curve plotted as absorbance vs. Trolox concentration.

3.5. DPPH Radical Scavenging Activity

The free radical scavenging activity was evaluated using the stable radical DPPH, according to the method described by Brand-Williams et al. with a slight modification [15]. For each compound, a 1:1 ratio expressed as moles of compound per mole of DPPH radical was tested. A 1 mL sample of the compound solution in ethanol was added to 1 mL of DPPH solution in ethanol so that the initial DPPH concentration in the cuvettes was 0.1 mM. The samples were incubated for 1 h at 20 °C in ethanol and the decrease in the absorbance values of the DPPH solution was measured at λmax 517 nm on the Evolution 300 UV-Visible (Thermo Scientific) cuvette spectrophotometer. Results were expressed as scavenging activity calculated as follows:
Scavenging activity, % = [(Ao− A1)/Ao] × 100
The concentration of compound needed to decrease 50% of the initial DPPH concentration (EC50) is a parameter widely used to measure the antioxidant effect. For determination of the EC50 the values of DPPH decrease after 1 h were used. The EC50 values were calculated graphically by plotting scavenging activity against compound concentration. Different sample concentrations (0.01, 0.02, 0.04, 0.06, 0.08 and 0.1 mM) were used in order to obtain kinetic curves and to calculate the EC50 values.

3.6. Lipoxygenase Inhibition Assessment

Lipoxygenase type 1B (LOX-1B) from Glycine max (soybean), boric acid, linoleic acid, ammonium acetate, CuCl2, and ethanol (96%) were purchased from Sigma-Aldrich and were used with no further purification. LOX-1B inhibition activity was determined spectrophotometrically [16] by measuring the increase in absorbance at 234 nm for the oxidation of linoleic acid on the Multiskan Go microplate spectrophotometer (Thermo Scientific). The reaction mixture contained 3 µL of test compounds dissolved in DMSO at initial concentrations of 0.05–2 mM (final concentrations 0.8, 3.3, 8.3, 16.6, 33.3 µM); 100 µL of 0.3 mM linoleic acid; 30 µL of borate buffer (pH = 9.0) and 17 µL of lipoxygenase solution in borate buffer. The total sample volume was 150 µL. The total cell volume used for UV-Vis measurements is 300 µL. The increase in absorbance was recorded every 10 s during 10 min under a controlled temperature 25 °C. All experiments were performed in triplicate.
The degree of LOX-1B activity (I %) in the presence of the complexes (Table 5) was calculated according to the following [5]:
I, % = (νo in the presence of inhibitor/νo in the absence of inhibitor) × 100,
where νo is the initial rate. The value of the initial rate (νo, µM min−1) was calculated according to the formula:
νo = ∆C/∆t = ∆A/∆tε = tgα/∆tε,
where C is the concentration of product (hydroperoxy-linoleic acid), t is the reaction time, ε is the molar absorbance coefficient of hydroperoxy-linoleic acid and tgα is the slope of the kinetic curve plotted as absorbance vs. time.

3.7. Antiproliferative Activity [28]

The human HCT116 colorectal carcinoma, A549 non-small cell lung carcinoma, MCF7 breast adenocarcinoma cell lines and WI-38 diploid human cell line composed of fibroblasts were obtained from the European collection of authenticated cell cultures (ECACC; Salisbury, UK) All cells were grown in a DMEM medium (Gibco™, Ireland) supplemented with 10% fetal bovine serum (Gibco™, Brazil). The cells were cultured in an incubator at 37 °C in a humidified 5% CO2 atmosphere and were sub-cultured 2 times a week. The effect of the investigated compounds on cell proliferation was evaluated using a common MTT assay. The cells were seeded in 96-well tissue culture plates («TPP», Trasadingen, Switzerland) at 7 × 103 cells/well in 100 µL of the medium. After overnight incubation at 37 °C, the cells were treated with the tested compounds in the concentration range of 0 to 100 µM. Cisplatin was used as a standard. After 72 h of treatment, the solution was removed, and a freshly diluted MTT solution (100 µL, 0.5 mg/mL in cell medium) was added to the wells, and the plates were further incubated for 50 min. Subsequently, the medium was removed, and the formazan product was dissolved in 100 μL of DMSO. The number of living cells in each well was evaluated by measuring the absorbance at 570 nm using the «Zenith 200 rt» microplate reader (Biochrom, Cambridge, UK) The meanings of 50% inhibition concentration (IC50) with standard deviation were calculated using GraphPad Prism Version 5.03 for Windows.

3.8. Cell Death and Cell Cycle Analysis

HCT-116 cells (colon carcinoma) (1 × 106) were seeded in a 6-well plate and incubated with 2, 5 and cisplatin (values based on MTT assay) at 2 × IC50 for 24 and 48 h. After incubation, the cells were harvested by trypsinization, precipitated by centrifugation (3500 rpm), washed with cold PBS and recentrifuged. Aliquots of cells were processed as recommended in the Muse AnnexinV&Dead Cell Kit. The results were recorded on a Muse Cell Analyzer flow cytometer (Luminex corp., Austin, TX, USA) [20].
For the cell cycle analysis, HCT-116 cells (1 × 106) were seeded in a six-well plate and were incubated for 24 h and then the medium was treated with compounds at the concentrations IC50 and incubated for 24 h. After incubation, the cells were harvested by trypsinization and precipitated by centrifugation (3500 rpm). After precipitation, the supernatant was removed, washed with PBS, centrifuged, fixed with 70% ethanol and incubated for at least 3 h at −20 °C. After incubation, 200 µL of the cell suspension was collected, centrifuged, the supernatant was removed, and washed with 200 µL of PBS. Then, the cells were stained 200 µL of the Muse Cell Cycle Reagent and incubated for 30 min at r.t. in the dark. Cell cycle analysis was performed using a Muse Cell Analyzer flow cytometer (Merck, Rahway, NJ, USA).

3.9. Tubulin Polymerization

Evaluation of the effect of compounds on tubulin polymerization performed according to [29]. A crude fraction of tubulin and microtubule-associated proteins (Tb+MAP) was isolated from mouse brain tissue by a polymerization–depolymerization method [30].
The procedure of mouse euthanasia using cervical dislocation for Tb+MAP preparation is in compliance with the Guidelines for Animal Experiments, which were approved by the local bioethics committee of the IPAC RAS. The freshly dissected brain was immediately placed on ice, cleared of meninges and surface blood vessels, and washed with cold buffer A (50 mM Tris-HCl, pH 6.9 at 4 °C, 2 mM EGTA). The extracted brain tissue was homogenized under ice cooling in the same buffer using a Potter S homogenizer (Sartorius, Göttingen, Germany). The resulting homogenate was centrifuged at 10,000× g for 30 min on an Avanti 25 centrifuge (Beckman, Brea, CA, USA) using a JA-14 rotor for the precipitation of non-ruptured cells. The precipitate was discarded, and the supernatant was again centrifuged at 100,000× g for 60 min at 4 °C on an Optima MAX XP centrifuge (Beckman, Brea, CA, USA) using a MLA-50 rotor. The precipitate was again discarded. The protein concentration in the supernatant was determined by the Bradford assay using the Bio-Rad Protein Assay kit (Bio-Rad, Hercules, CA, USA) according to the manufacturer’s instructions. The resulting supernatant was a cytosolic fraction enriched in microtubular proteins (Tb and MAP). The Tb+MAP preparation was centrifuged at 5000× g for 10 min at 4 °C immediately before polymerization in order to remove denatured and aggregated protein molecules by precipitation. The Tb+MAP polymerization was performed at 37 °C in buffer A in the presence of 0.1 mM GTP after the addition of 100 M of test compound or the same volume of vehicle (DMSO). The protein concentration in the sample was 0.2 mg/mL. The polymerization kinetics was monitored on a Victor 3 or EnVision microplate reader (Perkin Elmer, Waltham, MA, USA) at 355 nm.

4. Conclusions

Novel polyfunctional organotin complexes 15 based on a Schiff base containing an antioxidant 2,6-di-tert-butylphenol moiety were obtained and characterized. Using XRD method, the structures of initial ligand L and compound 2 were defined. In complex 2 the ligand is coordinated by O atom to the Sn center and distorted octahedron geometry was observed. In addition, a proton transfer from the oxygen atom to the nitrogen atom in the ligand part was revealed for complex 2.
Complexes 1 and 5 demonstrated high antioxidant activity in CUPRAC- and DPPH-methods due to their ability to be involved in one-electron and hydrogen atom transfer. The antioxidant activity was also explored in the enzymatic oxidation of linoleic acid by lipoxygenase (LOX 1-B) in vitro. Only compound 4 was found to be a moderate inhibitor of LOX 1-B.
The antiproliferative activity of complexes 15 was studied in vitro on HCT-116, MCF-7, A-549 and WI-38 cancer cell lines in the MTT test. It was established that dibutyl- and triphenyltin complexes 2 and 5 were the most active. Apoptosis and the cell cycle analysis were investigated by flow cytometry on HCT-116 cell line for these compounds after 24 and 48 h. Complex 2 was found to be the most active compound inducing apoptosis. Moreover, compounds 2 and 5 blocked proliferation in the G2/M phase of the cell cycle. The mechanism of antiproliferative activity of compounds 2 and 5 may be also connected with their influence on Tb+MAP polymerization.
Thus, the obtained compounds demonstrate high biological activity, which makes thempromising for further research as potential anti-tumor agents with possible combined actions.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24032024/s1.

Author Contributions

Conceptualization, T.A.A. and A.A.N.; Funding acquisition, T.A.A.; Investigation Y.A.G., D.B.S., M.A.V., D.M.M., V.A.T., Y.F.O., E.F.S., P.N.S. and A.A.N.; Supervision, E.R.M.; Writing—original draft preparation, T.A.A., Y.A.G., D.B.S., D.M.M. and E.R.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by financial support of scholarship of the President of the Russian Federation for young scientists and graduate students (Grants Council of the President of the Russian Federation for state support of young Russian scientists and state support of the leading scientific schools of the Russian Federation, No. SP-1641.2021.4).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Copies of 1H, 13C, 119Sn NMR, ECI-MS spectra and structure files of the synthesized compounds are presented in the Supplementary Materials.

Acknowledgments

The authors are thankful to Alexander Makarov for providing the Orbitrap Elite mass spectrometer for this work. We also acknowledge the use of a STOE STADIVARI single-crystal X-ray diffractometer purchased under the Lomonosov MSU Program of Development and the “Centre for Collective Use of IPAC RAS” (IPAC research topic FFSN-2021-0005) for use of equipment for biological assay of tubulin polymerization.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Milaeva, E.R.; Tyurin, V.Y. Hybrid metal complexes with opposed biological modes of action—Promising selective drug candidates. Pure Appl. Chem. 2017, 89, 1065–1088. [Google Scholar] [CrossRef]
  2. Milaeva, E.R.; Shpakovsky, D.B.; Gracheva, Y.A.; Antonenko, T.A.; Ksenofontova, T.D.; Nikitin, E.A.; Berseneva, D.A. Novel selective anticancer agents based on Sn and Au complexes. Mini-review. Pure Appl. Chem. 2020, 92, 1201–1216. [Google Scholar] [CrossRef]
  3. Antonenko, T.A.; Gracheva, Y.A.; Shpakovsky, D.B.; Vorobyev, M.A.; Tafeenko, V.A.; Mazur, D.M.; Milaeva, E.R. Cytotoxic activity of organotin compounds containing non-steroidal anti-inflammatory drugs. J. Organomet. Chem. 2022, 960, 122191. [Google Scholar] [CrossRef]
  4. Adeyemi, J.O.; Onwudiwe, D.C. Organotin(IV) Dithiocarbamate Complexes: Chemistry and Biological Activity. Molecules 2018, 23, 2571. [Google Scholar] [CrossRef] [Green Version]
  5. Xanthopoulou, M.N.; Hadjikakou, S.K.; Hadjiliadis, N.; Milaeva, E.R.; Gracheva, J.A.; Tyurin, V.Y.; Kourkoumelis, N.; Christoforidis, K.C.; Metsios, A.K.; Karkabounas, S.; et al. Biological studies of new organotin(IV) complexes of thioamide ligands. Eur. J. Med. Chem. 2008, 43, 327–335. [Google Scholar] [CrossRef]
  6. Dave, S.; Bansal, N. Analgesic And Anti-Inflammatory Activities Of Schif F Base Metal Complexes—A Review. Int. J. Basic App. Chem. Sci. 2013, 3, 31–40. [Google Scholar]
  7. Elshafie, H.S.; Sadeek, S.A.; Camele, I.; Mohamed, A.A. Biochemical Characterization of New Gemifloxacin Schiff Base (GMFX-o-phdn) Metal Complexes and Evaluation of Their Antimicrobial Activity against Some Phyto- or Human Pathogens. Int. J. Mol. Sci. 2022, 23, 2110. [Google Scholar] [CrossRef]
  8. Mohamed, A.A.; Ahmed, F.M.; Zordok, W.A.; El-Shwiniy, W.H.; Sadeek, S.A.; Elshafie, H.S. Novel Enrofloxacin Schiff Base Metal Complexes: Synthesis, Spectroscopic Characterization, Computational Simulation and Antimicrobial Investigation against Some Food and Phyto-Pathogens. Inorganics 2022, 10, 177. [Google Scholar] [CrossRef]
  9. El-Attar, M.S.; Ahmed, F.M.; Sadeek, S.A.; Mohamed, S.F.; Zordok, W.A.; El-Shwiniy, W.H. Characterization, DFT, and antimicrobial evaluation of some new N2O2 tetradentate Schiff base metal complexes. Appl. Organomet. Chem. 2022, 36, e6826. [Google Scholar] [CrossRef]
  10. Gielen, M.; Tiekink, E.R.T. (Eds.) Metallotherapeutic Drugs and Metal-Based Diagnostic Agents: The Use of Metals in Medicine; Wiley: Chichester, UK, 2008; p. 656. [Google Scholar]
  11. Shpakovsky, D.B.; Banti, C.N.; Mukhatova, E.M.; Gracheva, Y.A.; Osipova, V.P.; Berberova, N.T.; Albov, D.V.; Antonenko, T.A.; Aslanov, L.A.; Milaeva, E.R.; et al. Synthesis, antiradical activity and in vitro cytotoxicity of novel organotin complexes based on 2,6-di-tert-butyl-4-mercaptophenol. Dalton Trans. 2014, 43, 6880–6890. [Google Scholar] [CrossRef]
  12. Kolyada, M.N.; Osipova, V.P.; Berberova, N.T.; Shpakovsky, D.B.; Milaeva, E.R. Porphyrins with Phenolic Fragments at the Periphery of the Macrocycle as Perspective Antioxidants, Cytoprotectors and Heavy Metal Scavengers. Chem. Heterocycl. Compd. 2021, 57, 875–888. [Google Scholar] [CrossRef]
  13. Antonenko, T.A.; Shpakovsky, D.B.; Vorobyov, M.A.; Gracheva, Y.A.; Kharitonashvili, E.V.; Dubova, L.G.; Shevtsova, E.F.; Tafeenko, V.A.; Aslanov, L.A.; Iksanova, A.G.; et al. Antioxidative vs cytotoxic activities of organotin complexes bearing 2,6-di-tert-butylphenol moieties. Appl. Organomet. Chem. 2018, 32, e4381. [Google Scholar] [CrossRef]
  14. Apak, R.; Güçlü, K.; Özyürek, M.; Karademir, S.E. Novel Total Antioxidant Capacity Index for Dietary Polyphenols and Vitamins C and E, Using Their Cupric Ion Reducing Capability in the Presence of Neocuproine:  CUPRAC Method. J. Agric. Food. Chem. 2004, 52, 7970–7981. [Google Scholar] [CrossRef]
  15. Brand-Williams, W.; Cuvelier, M.E.; Berset, C. Use of a free radical method to evaluate antioxidant activity. LWT Food Sci. Technol. 1995, 28, 25–30. [Google Scholar] [CrossRef]
  16. Ozturk, I.; Filimonova, S.; Hadjikakou, S.K.; Kourkoumelis, N.; Dokorou, V.; Manos, M.J.; Tasiopoulos, A.J.; Barsan, M.M.; Butler, I.S.; Milaeva, E.R.; et al. Structural Motifs and Biological Studies of New Antimony(III) Iodide Complexes with Thiones. Inorg. Chem. 2010, 49, 488–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Nikš, M.; Otto, M. Towards an optimized MTT assay. J. Immunol. Methods 1990, 130, 149–151. [Google Scholar] [CrossRef]
  18. Hadjikakou, S.K.; Hadjiliadis, N. Antiproliferative and anti-tumor activity of organotin compounds. Coord. Chem. Rev. 2009, 253, 235–249. [Google Scholar] [CrossRef]
  19. Banti, C.N.; Hadjikakou, S.K.; Sismanoglu, T.; Hadjiliadis, N. Anti-proliferative and antitumor activity of organotin(IV) compounds. An overview of the last decade and future perspectives. J. Inorg. Biochem. 2019, 194, 114–152. [Google Scholar] [CrossRef]
  20. Vermes, I.; Haanen, C.; Steffens-Nakken, H.; Reutellingsperger, C. A novel assay for apoptosis Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. J. Immunol. Methods 1995, 184, 39–51. [Google Scholar] [CrossRef] [Green Version]
  21. Milaeva, E.R.; Shpakovsky, D.B.; Gracheva, Y.A.; Antonenko, T.A.; Osolodkin, D.I.; Palyulin, V.A.; Shevtsov, P.N.; Neganova, M.E.; Vinogradova, D.V.; Shevtsova, E.F. Some insight into the mode of cytotoxic action of organotin compounds with protective 2,6-di-tert-butylphenol fragments. J. Organomet. Chem. 2015, 782, 96–102. [Google Scholar] [CrossRef]
  22. Cheng, Z.; Lu, X.; Feng, B. A review of research progress of antitumor drugs based on tubulin targets. Transl. Cancer Res. 2020, 9, 4020–4027. [Google Scholar] [CrossRef] [PubMed]
  23. Medzhidov, A.A.; Kasumov, V.T.; Mamedov, K.S. Synthesis and study of chelate compounds of metals containing spatially hindered phenol. Koord. Khim. 1981, 7, 66–72. [Google Scholar]
  24. Stoe&Cie. X-AREA, X-RED32; Stoe&Cie: Darmstadt, Germany, 2012. [Google Scholar]
  25. Farrugia, L. WinGX and ORTEP for Windows: An update. J. Appl. Crystallogr. 2012, 45, 849–854. [Google Scholar] [CrossRef]
  26. Sheldrick, G. A short history of SHELX. Acta Crystallogr. Sect. A Found. Crystallogr. 2008, 64, 112–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Macrae, C.F.; Edgington, P.R.; McCabe, P.; Pidcock, E.; Shields, G.P.; Taylor, R.; Towler, M.; van de Streek, J. Mercury: Visualization and analysis of crystal structures. J. Appl. Crystallogr. 2006, 39, 453–457. [Google Scholar] [CrossRef] [Green Version]
  28. Shutkov, I.A.; Okulova, Y.N.; Tyurin, V.Y.; Sokolova, E.V.; Babkov, D.A.; Spasov, A.A.; Gracheva, Y.A.; Schmidt, C.; Kirsanov, K.I.; Shtil, A.A.; et al. Ru(III) Complexes with Lonidamine-Modified Ligands. Int. J. Mol. Sci. 2021, 22, 13468. [Google Scholar] [CrossRef]
  29. Bachurin, S.O.; Makhaeva, G.F.; Shevtsova, E.F.; Aksinenko, A.Y.; Grigoriev, V.V.; Shevtsov, P.N.; Goreva, T.V.; Epishina, T.A.; Kovaleva, N.V.; Pushkareva, E.A.; et al. Conjugation of Aminoadamantane and γ-Carboline Pharmacophores Gives Rise to Unexpected Properties of Multifunctional Ligands. Molecules 2021, 26, 5527. [Google Scholar] [CrossRef]
  30. Shelanski, M.L.; Gaskin, F.; Cantor, C.R. Microtubule assembly in the absence of added nucleotides. Proc. Natl. Acad. Sci. USA 1973, 70, 765–768. [Google Scholar] [CrossRef]
Scheme 1. Synthesis of organotin complexes 15 based on L.
Scheme 1. Synthesis of organotin complexes 15 based on L.
Ijms 24 02024 sch001
Figure 1. ESI mass spectrum of compound 1.
Figure 1. ESI mass spectrum of compound 1.
Ijms 24 02024 g001
Figure 2. Molecular structure of starting compound L.
Figure 2. Molecular structure of starting compound L.
Ijms 24 02024 g002
Figure 3. Molecular structure of 2 (a); hydrogen bonds in crystal packing 2 (b).
Figure 3. Molecular structure of 2 (a); hydrogen bonds in crystal packing 2 (b).
Ijms 24 02024 g003
Figure 4. Survival of cell lines HCT-116, MCF-7, A-549 and WI-38 in the presence of compounds 25.
Figure 4. Survival of cell lines HCT-116, MCF-7, A-549 and WI-38 in the presence of compounds 25.
Ijms 24 02024 g004
Figure 5. HCT-116 apoptotic profile after treatment with cisplatin and compounds 2, 5 after 24 h. Concentration of compounds 2·IC50 (µM).
Figure 5. HCT-116 apoptotic profile after treatment with cisplatin and compounds 2, 5 after 24 h. Concentration of compounds 2·IC50 (µM).
Ijms 24 02024 g005
Figure 6. HCT-116 apoptotic profile after treatment with cisplatin and compounds 2, 5 after 48 h. Concentration of compounds 2·IC50 (µM).
Figure 6. HCT-116 apoptotic profile after treatment with cisplatin and compounds 2, 5 after 48 h. Concentration of compounds 2·IC50 (µM).
Ijms 24 02024 g006
Figure 7. HCT-116 cell cycle analysis after treatment with compounds 2 and 5. Concentration of compounds IC50 (µM).
Figure 7. HCT-116 cell cycle analysis after treatment with compounds 2 and 5. Concentration of compounds IC50 (µM).
Ijms 24 02024 g007
Figure 8. The influence of compounds 2 and 5 (100 µM) on Tb+MAP polymerization. Results presented as mean ± SD obtained using at least from three independent experiments.
Figure 8. The influence of compounds 2 and 5 (100 µM) on Tb+MAP polymerization. Results presented as mean ± SD obtained using at least from three independent experiments.
Ijms 24 02024 g008
Table 1. Crystal data and the structure refinement details for compounds L and 2.
Table 1. Crystal data and the structure refinement details for compounds L and 2.
CompoundL2
Empirical formulaC21H27NO2C25H36ClNO2Sn0.50
Fw
T, K
325.43
293(2)
477.33
293(2)
Space groupP212121P21/n
SyngonyOrthorhombicMonoclinic
a (Å)6.0356(4)9.5216(5)
b (Å)17.5484(14)11.7902(7)
c (Å)17.7780(12)22.4464(11)
α (°)90.0090.00
β (°)90.0098.820(3)
γ (°)90.0090.00
V3)1883.0(2)2490.1(2)
Z
λ
4
MoKα
4
CuKα
Δρmax/Δρmin(e3)0.212/−0.2102.028/−1.732
μ (mm−1)0.0735.396
GOOF0.8641.122
R1/wR2(I ≥ 2σ(I)0.0458/0.11720.0772/0.2112
Table 2. Selected bond distances (Å) and angles (°) for compounds L and 2. Symmetry operation i = −x, 1 − y, 1 − z.
Table 2. Selected bond distances (Å) and angles (°) for compounds L and 2. Symmetry operation i = −x, 1 − y, 1 − z.
L2
Selected bond distances (Å)
O(1)-C(4)
O(2)-C(13)
N(1)-C(7)
N(1)-C(1)
1.365(3)
1.342(3)
1.272(3)
1.419(3)
Sn(1)-C(22)
Sn(1)-O(2)
Sn(1)-Cl(1)
2.140(7)
2.258(4)
2.6123(18)
Angles (°)
C(4)-O(1)-H(31)
C(13)-O(2)-H(32)
C(7)-N(1)-C(1)
C(7)-N(1)-H(32)
C(1)-N(1)-H(32)
C(6)-C(1)-N(1)
C(2)-C(1)-N(1)
O(1)-C(4)- C(5)
O(1)-C(4)-C(3)
N(1)-C(7)-C(8)
N(1) C(7)-H(7)
O(2)-C(13)-C(12)
O(2)-C(13)-C(8)
113(2)
103.9(17)
123.4(2)
101.4(14)
134.8(14)
116.1(2)
124.5(2)
115.2(2)
122.6(2)
121.8(2)
124.3(14)
119.6(2)
120.5(2)
C(22)-Sn(1)-O(2)
C(22)i-Sn(1)-O(2)
C(22)-Sn(1)-O(2)i
C(22)i-Sn(1)-O(2)i
O(2)-Sn(1)-O(2)i
C(22)-Sn(1)-Cl(1)
C(22)i -Sn(1)-Cl(1)
O(2)-Sn(1)-Cl(1)
O(2)i -Sn(1)-Cl(1)
C(22)-Sn(1)-Cl(1)i
C(22)i -Sn(1)-Cl(1)i
O(2)-Sn(1)-Cl(1)i
O(2)i -Sn(1)-Cl(1)i
88.31(19)
91.69(19)
91.69(19)
88.31(19)
180.00(18)
91.89(15)
88.11(15)
92.11(12)
87.89(12)
88.11(15)
91.89(15)
87.89(12)
92.11(12)
Table 3. The TEAC values of Cu2+ reducing activity in CUPRAC test for compounds L, 15.
Table 3. The TEAC values of Cu2+ reducing activity in CUPRAC test for compounds L, 15.
CompoundTEAC (CUPRAC)
L0.81 ± 0.03 [13]
13.37 ± 0.03
22.78 ± 0.04
31.19 ± 0.09
42.64 ± 0.07
52.31 ± 0.06
Results presented as mean ± SD obtained using data from at least three independent experiments.
Table 4. EC50 and k values in the DPPH test (EtOH, 20°C) in the presence of 15.
Table 4. EC50 and k values in the DPPH test (EtOH, 20°C) in the presence of 15.
Compound 12345
EC50, µM 16 ± 220 ± 330 ± 319 ± 223 ± 2
s 0.320.40.300.380.23
s−1 3.132.53.362.604.32
Ck (L mol−1 s−1)
0.06 mM*49 ± 5***
0.04 mM31 ± 429 ± 35 ± 149 ± 410 ± 1
0.02 mM 13 ± 42 ± 0.51 ± 0.23 ± 0.53 ± 0.4
0.01 mM1 ± 0.30.7 ± 0.10.9 ± 0.12 ± 0.10.7 ± 0.1
s = 2∙EC50/C0(DPPH)—the number of moles of compound required to reduce 1 mole of DPPH; s−1—the number of moles of DPPH reduced by 1 mole of the compound; the initial concentration of DPPH is 0.1 mM. Results presented as mean ± SD obtained using at least from three independent experiments.*—the parameter is not defined due to the high speed of the reaction.
Table 5. EC50 (μM) and degree of inhibition (I, %) of LOX 1-B lipoxygenase in the presence of compound 2.
Table 5. EC50 (μM) and degree of inhibition (I, %) of LOX 1-B lipoxygenase in the presence of compound 2.
C, μMI, %
0.822.66
8.327.59
16.631.21
33.339.55
Table 6. IC50 values (µM) against HCT-116, MCF-7, A-549 and WI-38 cell lines for compounds 15 and cisplatin.
Table 6. IC50 values (µM) against HCT-116, MCF-7, A-549 and WI-38 cell lines for compounds 15 and cisplatin.
Compound IC50, µM
HCT-116MCF-7A-549WI-38
158.2 ± 1480.6 ± 689 ± 1758.2 ± 8
21.6 ± 0.24.3 ± 0.55.7 ± 11.6 ± 0.2
33.8 ± 0.56.3 ± 0.77.7 ± 1.42.3 ± 0.3
43.1 ± 0.45.1 ± 0.66.8 ± 1.82.1 ± 0.3
50.32 ± 0.080.29 ± 0.030.30 ± 0.040.2 ± 0.05
cisplatin9.04 ± 0.711 ± 116.7 ± 34.8 ± 0.5
Results presented as mean ± SD obtained using at least from three independent experiments.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Antonenko, T.A.; Gracheva, Y.A.; Shpakovsky, D.B.; Vorobyev, M.A.; Mazur, D.M.; Tafeenko, V.A.; Oprunenko, Y.F.; Shevtsova, E.F.; Shevtsov, P.N.; Nazarov, A.A.; et al. Biological Activity of Novel Organotin Compounds with a Schiff Base Containing an Antioxidant Fragment. Int. J. Mol. Sci. 2023, 24, 2024. https://doi.org/10.3390/ijms24032024

AMA Style

Antonenko TA, Gracheva YA, Shpakovsky DB, Vorobyev MA, Mazur DM, Tafeenko VA, Oprunenko YF, Shevtsova EF, Shevtsov PN, Nazarov AA, et al. Biological Activity of Novel Organotin Compounds with a Schiff Base Containing an Antioxidant Fragment. International Journal of Molecular Sciences. 2023; 24(3):2024. https://doi.org/10.3390/ijms24032024

Chicago/Turabian Style

Antonenko, Taisiya A., Yulia A. Gracheva, Dmitry B. Shpakovsky, Mstislav A. Vorobyev, Dmitrii M. Mazur, Victor A. Tafeenko, Yury F. Oprunenko, Elena F. Shevtsova, Pavel N. Shevtsov, Alexey A. Nazarov, and et al. 2023. "Biological Activity of Novel Organotin Compounds with a Schiff Base Containing an Antioxidant Fragment" International Journal of Molecular Sciences 24, no. 3: 2024. https://doi.org/10.3390/ijms24032024

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop