Next Article in Journal
Electrochemical Performances of a Solid Oxide Electrolysis Short Stack Under Multiple Steady-State and Cycling Operating Conditions
Previous Article in Journal
A Structure and Magnetism Study of {MnII3MnIVLnIII3} Coordination Complexes with Ln = Dy, Yb
Previous Article in Special Issue
Crystal Structure and Anti-Proliferative and Mutagenic Evaluation of the Palladium(II) Complex of Deoxyalliin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Arene Ruthenium Complexes Specifically Inducing Apoptosis in Breast Cancer Cells

1
Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, Victor Babes 41, 400012 Cluj-Napoca, Romania
2
Institute of Chemistry, University of Neuchatel, Ave. de Bellevaux 51, 2000 Neuchatel, Switzerland
3
Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Marinescu 23, 400337 Cluj-Napoca, Romania
4
Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babeş-Bolyai University, 11 Arany Janos Str., 400028 Cluj-Napoca, Romania
*
Authors to whom correspondence should be addressed.
Inorganics 2024, 12(11), 287; https://doi.org/10.3390/inorganics12110287
Submission received: 4 October 2024 / Revised: 25 October 2024 / Accepted: 29 October 2024 / Published: 2 November 2024
(This article belongs to the Special Issue Noble Metals in Medicinal Inorganic Chemistry)

Abstract

:
Monocationic arene ruthenium complexes (RuL1RuL4) incorporating phenothiazinyl-hydrazinyl-thiazole ligands (L1L4) have been synthesized, characterized and evaluated as anticancer agents. Their cytotoxicity, antiproliferative activity and alteration of apoptotic gene expression were studied on three cancer cell lines, a double positive breast cancer cell line MCF-7 and two triple negative breast cancer cell lines Hs578T and MDA-MB-231. All arene ruthenium complexes were able to reduce the viability of the breast cancer cell lines, with the highest cytotoxicities being recorded for the [(p-cymene)RuL3Cl]+ (RuL3) complex on the MCF-7 (IC50 = 0.019 µM) and Hs578T cell lines (IC50 = 0.095 µM). In the double positive MCF-7 breast cancer cells, the complexes [(p-cymene)RuL1Cl]+ (RuL1) and [(p-cymene)RuL2Cl]+ (RuL2) significantly upregulated pro-apoptotic genes including BAK, FAS, NAIP, CASP8, TNF, XIAP and BAD, while downregulating TNFSF10. In the triple negative breast cancer cell line Hs578T, RuL1 reduced TNFSF-10 and significantly upregulated BAK, CASP8, XIAP, FADD and BAD, while complex RuL2 also increased BAK and CASP8 expression, but had limited effects on other genes. The triple negative MDA-MB-231 cancer cells treated with RuL1 upregulated NOD1 and downregulated p53, while RuL2 significantly downregulated p53, XIAP and TNFSF10, with minor changes in other genes. The significant alterations in the expression of key apoptotic genes suggest that such complexes have the potential to target cancer cells.

Graphical Abstract

1. Introduction

Human breast cancer, one of the most common types of cancer according to the World Health Organization statistics, has been diagnosed in more than 2.3 million women in 2022 [1]. Triple negative breast cancer (TNBC) is the most aggressive form [2], with patients exhibiting a heightened propensity for early metastasis compared with those with other types of breast cancer [3]. TNBC is defined by a deficiency in progesterone (PR) and estrogen receptors (ER), as well as in human epidermal growth factor receptor 2 (HER2) expression [4,5]. These factors collectively contribute to the TNBC’s distinctive biological features, which makes it challenging to treat with current drugs [6,7,8]. In comparison with TNBC, the double positive breast cancer (ER+/PR+) phenotype is linked to a greater prevalence among older patients, showing smaller tumors and offering a more favorable prognosis in comparison with the ER+/PR- phenotype [9,10,11].
Ruthenium complexes are promising chemotherapeutic agents, and several Ru(II) derivatives have entered clinical trials, with, however, variable outcomes [12,13]. Among biologically active Ru(II) complexes, the arene ruthenium family possesses interesting antitumor activity, showing different mechanisms. Some arene ruthenium complexes have the ability to bind DNA in a covalent or non-covalent manner [14,15], while others showed interactions with cytochrome c [16], as well as other biomolecules [17]. Such diversity in their targets and modes of action is linked to their facility to modify their structures and properties by the introduction of a wide range of ligands, thus tuning their cytotoxic effect. For example, against TNBC, arene ruthenium complexes incorporating the bidentate bis(3,5-dimethylpyrazol-1-yl)methane [18] or iminophosphorane ligands [19] showed a suppression of TNBC growth in vivo, while arene ruthenium complexes with α-amino acid ligands (methionine and tryptophan) were able to induce selective apoptosis of MDA-MB-231 cells through DNA interaction [20].
In a previous study, we reported the synthesis and biological activity of hydrazinyl-thiazolo arene ruthenium complexes [21]. All complexes were more active than cisplatin and oxaliplatin on cervical (HeLa) and ovarian (A2780 and A2780cisR) cancer cells [21], and in vivo, the complexes were well tolerated after oral and intraperitoneal administrations in rats without altering the hematological profile [22]. Moreover, considering the antitumor activity of phenothiazinyl-hydrazinyl-thiazole derivatives on hepatic HepG2 and colorectal CC531S cells [23], we decided to extend our investigation on a new family of arene ruthenium complexes (RuL1RuL4) with phenothiazinyl-hydrazinyl-thiazolo ligands (L1L4) and to evaluate their antiproliferative activity against double positive (MCF-7) and triple negative (Hs578T, MDA-MB-231) breast cancer cell lines, as well as non-cancerous breast cells (fR2).

2. Results

The monocationic arene ruthenium complexes (RuL1RuL4) were synthesized by reacting [(p-cymene)RuCl2]2 with two equivalents of phenothiazinyl-hydrazinyl-thiazole derivatives (L1L4) in methanol at room temperature (Scheme 1). All complexes were isolated as their chloride salts in good yield, as racemic mixtures. Accordingly, the complexes showed the characteristic diastereotopic protons of the p-cymene group in the 1H NMR spectra, which was emphasized by the presence of four doublets between 4 and 5 ppm (aromatic protons of p-cymene) and two doublets around 1 ppm (methyl groups of the isopropyl moiety). In addition to the p-cymene signals, a triplet and a quadruplet can be associated with the N-ethyl group of the phenothiazine moiety at 1.5 and 4.0 ppm, respectively. In the ESI-MS spectra (positive mode), the parent signal is always associated with the cationic complex [(p-cymene)RuLCl]+ after the loss of the counteranion.
Then, the antiproliferative activity of all complexes was determined on various cell lines using MTT assays, the NBC cell lines (MDA-MB-231, Hs578T and MCF-7) as well as the non-cancerous breast cells (fR2). The complexes were initially dissolved in DMSO, and stock solutions of 1.0 M concentration were used to prepare biological solutions (concentrations ranging from 10 nM to 104 nM) for the in vitro tests, thus never exceeding 1% of DMSO. The stability of the complexes under physiological conditions was not studied in detail, however, aquation cannot be excluded [24], and the chelating ligand (L) is more strongly coordinated than the chloride, thus ensuring that the active complexes have an arene ruthenium phenothiazinyl-hydrazinyl-thiazolo structure. All cell lines were incubated for 24 h with the complexes RuL1RuL4. The MTT values after 24 h incubation expressed as % of control according to the log(concentration, nM) were determined (Figure 1). The IC50 values for each compound are presented in Table 1.
The MTT assays show that all complexes reduced the viability of breast cancer cell lines after 24 h of incubation at low μM concentrations. For the MCF-7 and MDA-MB-231 cancer cell lines, complexes RuL1 and RuL2 showed cell viability below 35% compared with the control experiment (cells incubated in the absence of complexes), while for the Hs578T cell lines, only RuL2 reduced cell viability below 15%, with the other complexes having a lower inhibitory activity. Based on these MTT assays, the IC50 values for each complex were determined (Table 1).
The lowest IC50 value was recorded for RuL3 (0.019 µM) on the double positive breast cancer cell line MCF-7, followed by the same complex on the triple negative breast cancer cell line Hs578T (0.095 µM). This complex showed the highest cytotoxicity among all complexes investigated but remained less cytotoxic on the triple negative breast cancer cell line MDA-MB-231 (14.125 µM). On the other hand, RuL2 showed a high degree of cytotoxicity on all cancer cell lines, MCF-7 (0.654 µM), MDA-MB-231 (0.449 µM) and Hs578T (0.705 µM), compared with RuL1, but was less cytotoxic than RuL3 on the MCF-7 and Hs578T breast tumor cell lines. Overall, for complexes RuL1 and RuL2, the IC50 values were much more homogenous and showed a superior correlation coefficient (R2), thus encouraging us to select RuL1 and RuL2 for further investigations.
The MTT results showed a decrease in cell proliferation, which could be associated with an increase in the number of apoptotic cells. Therefore, we evaluated the apoptosis rate of normal and breast cancer cell lines exposed to RuL1 and RuL2, at their respective IC50 concentrations (Figure 2). Twenty-four hours after treatment, cell death was assessed via fluorescence microscopy after staining with DAPI (4′,6-diamidino-2-phenylindole), a dye that specifically stains cell nuclei. As shown in Figure 2A, DAPI staining of the control group displayed intact and round nuclei, indicating that cells were not undergoing apoptosis. In treated cells, the number of viable cells decreased and fragmented nuclei were observed. In addition, both compounds exhibited visible pro-apoptotic effects on breast cell lines at 24 h post-treatment. Interestingly, RuL2 showed a higher percentage of apoptotic cells than RuL1 (Figure 2B), with the most pronounced effect observed in fR2 cells for RuL2, being in agreement with the MTT data.
We also evaluated the mode of action of RuL1 and RuL2 at the IC50 concentrations. Based on the MTT and apoptosis results, we selected for the qRT-PCR (real-time quantitative reverse transcription PCR) genes, which were strongly involved in apoptotic processes including TNFSF10 (Tumor Necrosis Factor Superfamily Member 10), NOD1 (Nucleotide Binding Oligomerization Domain Containing 1), CASP8 (Caspase 8), FADD (Fas Associated via Death Domain), NAIP (NLR Family Apoptosis Inhibitory Protein), FAS (Fas Cell Surface Death Receptor), BAD (BCL2 Associated Agonist of Cell Death), p53 (tumor protein p53), TNF (Tumor Necrosis Factor), BAK1 (BCL2-antagonist/killer 1) and XIAP (X-Linked Inhibitor of Apoptosis).
In the normal cell line fR2, treatment with RuL1 and RuL2 led to specific changes in apoptotic gene expression, with RuL1 causing downregulation of CASP8 and p53, while RuL2 upregulated BAK; however, changes in NAIP, FAS, FADD and TNFSF10 were not statistically significant. In double positive MCF-7 cells, both complexes significantly increased the levels of pro-apoptotic genes including BAK, FAS, NAIP, CASP8, TNF, XIAP and BAD, although changes in p53, FADD and NOD1 lacked statistical significance, and TNFSF10 expression decreased without statistical relevance. For triple negative Hs578T cells, RuL1 reduced TNFSF10 expression and enhanced levels of BAK, CASP8, XIAP, FADD, BAD and TNF, while RuL2 similarly increased BAK, NAIP, CASP8 and TNF but caused only a slight and non-significant decrease in FAS expression. In triple negative MDA-MB-231 cells, RuL1 treatment resulted in increased NOD1 and decreased p53 expression, with other genes showing non-significant changes, whereas RuL2 significantly lowered p53, XIAP and TNFSF10 levels, with slight upregulation of BAK and non-significant alterations in NAIP, TNF, FADD and BAD.
In the case of the triple negative breast cancer cell line Hs578T, we observed that RuL1 inhibited the relative gene expression of TNFSF10 in comparison with the control group. In addition, the expression levels of BAK1, CASP8, XIAP, FADD, BAD and TNF were statistically overexpressed compared with the control group. In the case of the FAS gene, the expression level was slightly decreased but not statistically significant. Otherwise, gene expression profiling in RuL2 was statistically overexpressed compared with control (Table 2, Figure 3). The second triple negative breast cancer cell line, MDA-MB-231, treated with RuL1, did not alter the expression levels of the selected genes. The NOD1 gene exhibited a statistically upregulated level, while the p53 gene exhibited a statistically downregulated level compared with the control group. BAK and TNF genes showed an increased expression level and XIAP, FADD, TNFSF10 and BAD and a decreased profiling with no significant p values. RuL2 statistically decreased significantly the expression of p53, XIAP and TNFSF10. However, RuL2 slightly upregulated the BAK1 level and respectively decreased the NAIP, TNF, FADD and BAD levels, with no significant p values compared with the control group (Table 2, Figure 3).

3. Discussion

Ruthenium complexes trigger cell-specific responses with variable toxicity and subsequent cell fate depending on the type of breast cancer cells. The ruthenium complexes, in particular RuL3 and RuL2, showed potent cytotoxic effects against various breast cancer cell lines, with RuL3 showing the highest cytotoxicity in both MCF-7 and Hs578T cells. The mechanism of action involves the induction of apoptosis, with variations in apoptotic gene expression profiles across the cell lines. This highlights the complex nature of ruthenium interactions with cancer cells.
The treatment of breast cancer cell lines with ruthenium complexes RuL1 and RuL2 resulted in significant alterations in the expression of key apoptotic genes, reflecting their potential as therapeutic agents. In normal fR2 cells, RuL1 and RuL2 predominantly affected CASP8 and p53, while RuL2 also upregulated BAK. In the MCF-7 breast carcinoma cell line, both complexes notably increased the expression of pro-apoptotic genes such as BAK, FAS, NAIP, CASP8, TNF, XIAP and BAD, while reducing TNFSF10 levels. Similarly, in the triple negative breast cancer cell line Hs578T, RuL1 decreased TNFSF10 and significantly upregulated BAK, CASP8, XIAP, FADD and BAD, while RuL2 also enhanced BAK and CASP8 expression but had variable effects on the other genes. MDA-MB-231 cells treated with RuL1 showed upregulation of NOD1 and downregulation of p53, while RuL2 led to significant decreases in p53, XIAP and TNFSF10, with minor changes in other genes. These findings highlight the ability of ruthenium complexes incorporating phenothiazinyl-hydrazinyl-thiazolo ligands to modulate apoptotic pathways in a cell type-specific manner, confirming their potential utility in targeted cancer therapy.

4. Materials and Methods

All chemicals, reagents and solvents were purchased from commercial suppliers and used without further purification unless otherwise stated. The 1H NMR spectra were recorded in solution (CDCl3) at room temperature on a Bruker Avance II 400 spectrometer, using an internal standard. Elemental analyses were performed by the Mikroelementarisches Laboratorium, ETH Zürich. Mass spectra (electrospray ionization, positive-ion mode) were recorded with a Bruker FTMS 4.7T BioAPEX II mass spectrometer. The phenothiazinyl-hydrazinyl-thiazole derivatives (L1L4) were prepared according to published method [23].

4.1. Synthesis and Characterization of Compounds RuL1RuL4

The dinuclear complex [(p-cymene)RuCl2]2 (100 mg; 0.163 mmol) and the corresponding L1L4 phenothiazinyl-hydrazinyl-thiazole (0.327 mmol) were dissolved in methanol (50 mL) and stirred at room temperature for 10 h. Then, the solvent was removed under vacuum. The residue was dissolved in dichloromethane (5 mL), and 200 mL of n-hexane was added to initiate the precipitation of the salts. The precipitate was filtered, washed several times with n-hexane, and dried under vacuum to give the corresponding product in good yield.
RuL1: [(p-cymene)RuL1Cl]Cl, yellow-green powder, yield 75%. 1H NMR (CDCl3, 400 MHz): δ = 1.02 (d, 3JH-H = 6.6 Hz, 3H), 1.07 (d, 3JH-H = 6.6 Hz, 3H), 1.52 (t, 3JH-H = 7.0 Hz, 3H), 2.25 (s, 3H), 2.50 (sept, 3JH-H = 6.6 Hz, 1H), 4.03 (q, 3JH-H = 7.0 Hz, 2H), 4.05 (d, 3JH-H = 5.7 Hz, 1H), 4.75 (d, 3JH-H = 5.7 Hz, 1H), 4.82 (d, 3JH-H = 5.7 Hz, 1H), 4.86 (d, 3JH-H = 5.7 Hz, 1H), 6.76 (s, 1H), 6.98–7.01 (m, 3H), 7.20–7.22 (m, 2H), 7.58–7.60 (m, 3H), 7.83 (m, 1H), 7.97–7.98 (m, 2H), 8.32 (s, 1H), 9.14 (s, 1H), 15.35 (s, 1H) ppm. IR (KBr pellet): 2918 (m), 1575 (m), 1466 (s), 1384 (m), 1243 (m), 1053 (w), 752 (w) cm−1. ESI-MS m/z (+): 699.1 [M-Cl]+. Anal. Calcd for C34H34N4S2Cl2Ru: C, 55.65; H, 4.53; N, 7.64; S, 8.74. Found: C, 55.23; H, 4.33; N, 7.56; S, 8.64.
RuL2: [(p-cymene)RuL2Cl]Cl, yellow-orange powder, yield 77%. 1H NMR (CDCl3, 400 MHz): δ = 1.12 (d, 3JH-H = 6.7 Hz, 3H), 1.21 (d, 3JH-H = 6.7 Hz, 3H), 1.52 (t, 3JH-H = 6.8 Hz, 3H), 2.35 (s, 3H), 2.46 (s, 3H), 2.86 (s, 3H), 3.18 (sept, 3JH-H = 6.7 Hz, 1H), 4.03 (q, 3JH-H = 6.8 Hz, 2H), 4.94 (d, 3JH-H = 5.9 Hz, 1H), 5.27 (d, 3JH-H = 5.9 Hz, 1H), 5.47 (d, 3JH-H = 5.9 Hz, 1H), 5.58 (d, 3JH-H = 5.9 Hz, 1H), 6.97–6.98 (m, 4H), 7.18–7.19 (m, 2H), 8.25 (s, 1H), 9.06 (s, 1H) ppm, 15.23 (s, 1H) ppm. IR (KBr pellet): 2921 (m), 1624 (s), 1468 (s), 1245 (m), 1110 (m), 751 (m) cm−1. ESI-MS m/z (+): 679.1 [M-Cl]+. Anal. Calcd for C31H34N4OS2Cl2Ru: C, 52.17; H, 4.66; N, 7.85; S, 8.99. Found: C, 52.12; H, 4.58; N, 7.71; S, 8.89.
RuL3: [(p-cymene)RuL3Cl]Cl, yellow-green powder, yield 68%. 1H NMR (CDCl3,400 MHz): δ = 1.08 (d, 3JH-H = 6.5 Hz, 3H), 1.18 (d, 3JH-H = 6.5 Hz, 3H), 1.48 (t, 3JH-H = 6.7 Hz, 3H), 2.34 (s, 3H), 2.46 (s, 3H), 2.65 (sept, 3JH-H = 6.5 Hz, 1H), 3.99 (q, 3JH-H = 6.7 Hz, 2H), 4.93 (d, 3JH-H = 5.6 Hz, 1H), 5.22 (m, 2H), 5.47 (d, 3JH-H = 5.6 Hz, 1H), 6.46 (s, 1H), 6.95–6.96 (m, 4H), 7.16–7.17 (m, 2H), 8.25 (s, 1H), 9.11 (s, 1H), 15.02 (s, 1H) ppm. IR (KBr pellet): 2921 (m), 1623 (s), 1466 (s), 1242 (m), 1136 (m), 753 (m) cm−1. ESI-MS m/z (+): 637.1 [M-Cl]+. Anal. Calcd for C29H32N4S2Cl2Ru: C, 51.86; H, 4.65; N, 8.34; S, 9.55. Found: C, 51.77; H, 4.57; N, 8.24; S, 9.43.
RuL4: [(p-cymene)RuL4Cl]Cl, yellow-orange powder, yield 82%. 1H NMR (CDCl3, 400 MHz): δ = 1.09 (d, 3JH-H = 6.7 Hz, 3H), 1.18 (d, 3JH-H = 6.7 Hz, 3H), 1.33 (t, 3JH-H = 7.1 Hz, 3H), 1.49 (t, 3JH-H = 6.9 Hz, 3H), 2.37 (s, 3H), 2.62 (sept, 3JH-H = 6.7 Hz, 1H), 2.82 (s, 3H), 4.00 (q, 3JH-H = 6.9 Hz, 2H), 4.30 (q, 3JH-H = 7.1 Hz, 2H), 4.93 (d, 3JH-H = 5.6 Hz, 1H), 5.24 (m, 2H), 5.47 (d, 3JH-H = 5.6 Hz, 1H), 6.96–6.97 (m, 4H), 7.14–7.15 (m, 2H), 8.25 (s, 1H), 9.08 (s, 1H), 15.92 (s, 1H) ppm. IR (KBr pellet): 2969 (m), 1599 (s), 1465 (s), 1372 (m), 1271 (m), 1098 (s), 753 (m) cm−1. ESI-MS m/z (+): 709.1 [M-Cl]+. Anal. Calcd for C32H36N4O2S2Cl2Ru: C, 51.68; H, 4.74; N, 7.53; S, 8.62. Found: C, 50.05; H, 4.68; N, 7.41; S, 8.55.

4.2. Cell Lines and Cell Culture

Two human triple negative breast cancer cell lines, MDA-MB-231 and Hs578T, a double positive breast cancer cell line, MCF-7, and a normal breast cell line, fR2, were purchased from the American Type Culture Collection (ATCC). MDA-MB-231 cells were cultured in RPMI-1640 medium (Gibco) supplemented with 10% fetal bovine serum (FBS, Gibco) and 1% penicillin-streptomycin (Gibco). The Hs578T cell line was maintained in D-MEM high glucose supplemented (Gibco) with 10% FBS, 1% penicillin-streptomycin, 2 mM L-glutamine (Gibco), 0.01 mg/mL insulin, 1% MEM-NEEA (MEM Non-Essential Amino Acids Solution, Gibco) and 1% penicillin-streptomycin. The MCF-7 cell line was cultured in MEM (Minimum Essential Medium Eagle, Gibco) supplemented with 10% FBS, 2 mM L-glutamine, 1% MEM-NEEA and 1% penicillin-streptomycin. Normal breast cells, fR2, were cultured in RPMI-1640, 10% FBS, 0.01 mg/mL insulin and 1% penicillin-streptomycin. The cells were grown in a humidified atmosphere at 37 °C with 5% CO2.

4.3. Cell Viability Assay

Cell viability was assessed by the MTT assay (MTT = 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) in accordance with the manufacturer’s protocol. At a seeding density of 104 cells/well, cells were seeded in a 96-well plate and incubated at 37 °C with 5% CO2. Twenty-four hours after incubation, the cells were treated with the appropriate therapeutic dose of the RuL1RuL4 and incubated for 24 h. The complexes were initially dissolved in DMSO, and stock solutions of 1.0 M concentration were used to prepare the biological solutions (concentrations ranging from 10 nM to 104 nM). To evaluate the proliferative activity of the cells, 1 mg/mL MTT solution was added and incubated for 1 h. The formazan obtained after intracellular reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide was spectrophotometrically quantified by dissolving formazan in dimethyl sulfoxide and measuring the absorbance after 15 min using a plate reader at a wavelength of 570 nm.

4.4. DAPI Staining

Nucleus traits were assessed using DAPI staining, 104 cells/well were seeded and treated with RuL1 or RuL2 24 h later, and after incubation, DAPI solution was added to each well and incubated for a further 5 min at 37 °C. After rinsing with 1X PBS, fluorescence microscopy was used to visualize nucleus features.

4.5. RNA Extraction, cDNA Synthesis and RT-PCR

Cells (seeding density of 5 × 105 cells/well) were treated with the appropriate IC50 doses of RuL1RuL4, determined for each cell line, and incubated for 24 h. Total RNA was extracted from all cell lines using TriReagent (Invitrogen) in agreement with the producer’s procedure. RNA concentration and quality were assessed using a Nanodop-1000 spectrophotometer (Thermo Scientific, Waltham, MA, USA). The amount of 1000 ng of total RNA was reversed transcribed into cDNA using a Transcriptor First Strand cDNA Synthesis Kit (Roche, Basel, Switzerland) based on the producer’s guidelines.
The gene expression evaluation was conducted using a TaqMan Fast Advanced Master Mix (Applied Biosystems, Waltham, MA, USA) based on the producer’s guidelines and qRT-PCR was performed on a ViiA7 System (Applied Biosystems). Each sample was analyzed in duplicate in a 10 µL volume using a 384-well plate.
The internal standard controls used were 18S and GAPDH rRNA, and the genes evaluated were BAK, FAS, NAIP, CASP8, TNF, p53, NOD1, XIAP, FACC, TNFSF10 and BAD. Data analysis was performed using the Qiagen online tool based on the ΔΔCt method. (https://www.qiagen.com/us/service-and-support/learning-hub/technologies-and-research-topics/rna-universe/gene-expression/data-analysis/, accessed on 1 November 2022).
TNFSF10: right CAGAGCCTTTTCATTCTTGGA: left CCTCAGAGAGTAGCAGCTCACA
NOD1: right GGCGAGATACTTCCCTCCTT: left GAATGCAAAGGCCTCACG
CASP8: right TTTCTGCTGAAGTCCATCTTTTT: left TAGGGGACTCGGAGACTGC
FADD: right AGGTCTAGGCGCTCTGC: left CCGAGCTCAAGTTCCTATGC
NAIP: right TGGGAGAATCCTCTCGTCAGA: left CTGGCCAGCATTCTCCTCTA
FAS: right GAGACGAAGCTCACGAAAAGC: left GGCCAAGTTGCTGAATCAAT
BAD: right GCTTCCTCTCCCACCGTAG: left ACCCGGAGACAGATGAG
p53: left TCAACAAGATGTTTTGCCAACTG: right TGTGCTGTGACTGCTTGTAGATG
TNF right GGGGAACTCTTCCCTCTG: left CAGCCTCTTCTCCTTCCTGAT
BAK1: right CCGCGAGACTCCAGTGAT: left GGCCCACAGAGCAACTTCC
GAPDH: right CACCTTCCCCATGGTGTCT: left CCCCGGTTTCTATAAATTGAGC
18S: right CGCTCCACCAACTAAGAACG: left CTCAACACGGGAAACCTCAC

4.6. Statistical Analysis

Results are reported as mean ± SD (standard deviation). The differences between the analyzed RuL1RuL4 activity on cancer cells and controls were analyzed using the t-test (p ≤ 0.005 was considered statistically significant). Statistical analyses were performed using GraphPad Prism version 6 software [26] and STRING 8.0 free version [25].

5. Conclusions

Four new monocationic arene Ru(II) complexes with bidentate phenothiazinyl-hydrazinyl-thiazolo ligands were prepared and their cytotoxicity, antiproliferative activity and alteration of apoptotic gene expression were investigated on three cancer cell lines, a double positive breast cancer cell line, MCF-7, and two triple negative breast cancer cell lines, Hs578T and MDA-MB-231. Two complexes, RuL1 and RuL2, have shown significant alterations in the expression of key apoptotic genes, highlighting the ability of such complexes to modulate apoptotic pathways in a cell type-specific manner, thus confirming their potential as anticancer agents. Future research should focus on elucidating the detailed mechanisms of cellular internalization in view of generating ruthenium complexes with enhanced selectivity and efficacy toward tumor cells, while minimizing off-target effects on normal cells.

Author Contributions

Conceptualization, A.G. and C.B.; methodology, A.G. and B.T.; validation, I.B.-N., L.G. and A.G.; formal analysis, O.C., T.C., A.G. and C.B; investigation, A.G., T.C., A.J., L.R. and O.Z.; resources, B.T., I.B.-N. and L.G.; data curation, B.T.; writing—original draft preparation, B.T., L.G.; writing—review and editing, B.T.; supervision, B.T. and A.G.; project administration, B.T. and A.G.; funding acquisition, B.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Swiss Enlargement Contribution in the framework of the Romanian-Swiss Research Program, project number IZERZO-142198/1.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors on request.

Acknowledgments

Luiza Gaina wishes to thank the “Agence Universitaire de la Francophonie” project AUF DRECO-7867_SER-ECO for its support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. De Cicco, P.; Catani, M.V.; Gasperi, V.; Sibilano, M.; Quaglietta, M.; Savini, I. Nutrition and Breast Cancer: A Literature Review on Prevention, Treatment and Recurrence. Nutrients 2019, 11, 1514. [Google Scholar] [CrossRef] [PubMed]
  2. Yang, F.-W.; Mai, T.-L.; Lin, Y.-C.J.; Chen, Y.-C.; Kuo, S.-C.; Lin, C.-M.; Lee, M.-H.; Su, J.-C. Multipathway regulation induced by 4-(phenylsulfonyl)morpholine derivatives against triple-negative breast cancer. Arch. Pharm. Chem. Life Sci. 2024, 357, 2300435. [Google Scholar] [CrossRef] [PubMed]
  3. Ciocan-Cartita, C.A.; Jurj, A.; Zanoaga, O.; Cojocneanu, R.; Pop, L.; Moldovan, A.; Moldovan, C.; Zimta, A.A.; Raduly, L.; Pop-Bica, C.; et al. New insights in gene expression alteration as effect of doxorubicin drug resistance in triple negative breast cancer cells. J. Exp. Clin Cancer Res. 2020, 39, 241. [Google Scholar] [CrossRef] [PubMed]
  4. Park, S.-Y.; Choi, J.-H.; Nam, J.-S. Targeting Cancer Stem Cells in Triple-Negative Breast Cancer. Cancers 2019, 11, 965. [Google Scholar] [CrossRef]
  5. Keskinkılıc, M.; Gökmen-polar, Y.; Badve, S.S. Triple Negative Breast Cancers: An Obsolete Entity ? Clin. Breast Cancer 2024, 24, 1–6. [Google Scholar] [CrossRef]
  6. Braicu, C.; Pileczki, V.; Pop, L.; Petric, R.C.; Chira, S.; Pointiere, E.; Achimas-Cadariu, P.; Berindan-Neagoe, I. Dual Targeted Therapy with p53 siRNA and Epigallocatechingallate in a Triple Negative Breast Cancer Cell Model. PLoS ONE 2015, 10, e0120936. [Google Scholar] [CrossRef]
  7. Braicu, C.; Chiorean, R.; Irimie, A.; Chira, S.; Tomuleasa, C.; Neagoe, E.; Paradiso, A.; Achimas-Cadariu, P.; Lazar, V.; Berindan-Neagoe, I. Novel insight into triple-negative breast cancers, the emerging role of angiogenesis, and antiangiogenic therapy. Expert Rev. Mol. Med. 2016, 18, e18. [Google Scholar] [CrossRef]
  8. Pop, L.; Braicu, C.; Budisan, L.; Morar, G.B.; Monroig-Bosque, P.C. Double gene siRNA knockdown of mutant p53 and TNF induces apoptosis in triple-negative breast cancer cells. OncoTargets Ther. 2016, 9, 6921–6933. [Google Scholar] [CrossRef]
  9. Ng, C.; Pathy, N.B.; Taib, N.A.; Ho, G.; Mun, S.; Rhodes, A.; Looi, L.-M.; Yip, C.-H. Do Clinical Features and Survival of Single Hormone Receptor Positive Breast Cancers Differ from Double Hormone Receptor Positive Breast Cancers ? Asian Pac. J. Cancer Prev. 2014, 15, 7959–7964. [Google Scholar] [CrossRef]
  10. Gamrani, S.; Boukansa, S.; Benbrahim, Z.; Mellas, N.; Alaoui, F.F.; Melhouf, M.A.; Bouchikhi, C.; Banani, A.; Boubbou, M.; Bouhafa, T.; et al. The Prognosis and Predictive Value of Estrogen Negative/Progesterone Positive (ER−/PR+) Phenotype: Experience of 1159 Primary Breast Cancer from a Single Institute. Breast J. 2022, 2022, 9238804. [Google Scholar] [CrossRef]
  11. Wei, S. Hormone receptors in breast cancer: An update on the uncommon subtypes. Pathol. Res. Pract. 2023, 250, 154791. [Google Scholar] [CrossRef] [PubMed]
  12. Kannan, N.; Radhakrishnan, V.; Sinha, A. Unveiling the anticancer activity of ruthenium and iron complexes. Inorg. Chem. Commun. 2024, 165, 112512. [Google Scholar] [CrossRef]
  13. Swaminathan, S.; Haribabu, J.; Kavembu, R. From concept to Cure: The Road Ahead for Ruthenium-Based Anticancer Drugs. ChemMedChem 2024, e202400435. [Google Scholar] [CrossRef] [PubMed]
  14. Ganeshpandian, M.; Loganathan, R.; Suresh, E.; Riyasdeen, A.; Akbarsha, M.A.; Palaniandavar, M. New ruthenium(II) arene complexes of anthracenyl-appended diazacycloalkanes: Effect of ligand intercalation and hydrophobicity on DNA and protein binding and cleavage and cytotoxicity. Dalton Trans. 2014, 43, 1203–1219. [Google Scholar] [CrossRef]
  15. Kasim, M.; Subarkhan, M.; Ramesh, R. Ruthenium(II) arene complexes containing benzhydrazone ligands: Synthesis, structure and antiproliferative activity. Inorg. Chem. Front. 2016, 3, 1245–1255. [Google Scholar] [CrossRef]
  16. Biancalana, L.; Pratesi, A.; Chiellini, F.; Zacchini, S.; Funaioli, T.; Gabbiani, C.; Marchetti, F. Ruthenium Arene Complexes with Triphenylphosphane Ligands: Cytotoxic Activity Towards Pancreatic Cancer Cells, Interaction with Model Proteins, and Critical Effect of Ethacrynic Acid Substitution. New J. Chem. 2017, 41, 14574–14588. [Google Scholar] [CrossRef]
  17. Mannancherril, V.; Therrien, B. Strategies toward the enhanced permeability and retention effect by increasing the molecular weight of arene ruthenium metallaassemblies. Inorg. Chem. 2018, 57, 3626–3633. [Google Scholar] [CrossRef]
  18. Montani, M.; Badillo, G.V.; Hysi, A.; Lupidi, G.; Pettinari, R.; Gambini, V.; Tilio, M.; Marchetti, F.; Pettinari, C.; Ferraro, S.; et al. The water soluble ruthenium(II) organometallic compound [Ru(p-cymene)(bis(3,5-dimethylpyrazol-1-yl)methane)Cl]Cl suppresses triple negative breast cancer growth by inhibiting tumor infiltration of regulatory T cells. Pharmacol. Res. 2016, 107, 282–290. [Google Scholar] [CrossRef]
  19. Nayeem, N.; Sauma, S.; Ahad, A.; Rameau, R.; Kebadze, S.; Bazett, M.; Park, B.J.; Casaccia, P.; Prabha, S.; Hubbard, K.; et al. Insights into Mechanisms and Promising Triple Negative Breast Cancer Therapeutic Potential for a Water-Soluble Ruthenium Compound. ACS Pharmacol. Transl. Sci. 2024, 7, 1364–1376. [Google Scholar] [CrossRef]
  20. Mello-andrade, F.; Guedes, A.P.M.; Pires, W.C.; Velozo-s, V.S.; Delmond, K.A.; Mendes, D.; Molina, M.S.; Matuda, L.; Montes de Sousa, M.A.; Melo-Reis, P.; et al. Ru(II)/amino acid complexes inhibit the progression of breast cancer cells through multiple mechanism-induced apoptosis. J. Inorg. Biochem. 2022, 226, 111625. [Google Scholar] [CrossRef]
  21. Grozav, A.; Balacescu, O.; Balacescu, L.; Cheminel, T.; Berindan-Neagoe, I.; Therrien, B. Synthesis, Anticancer Activity, and Genome Profiling of Thiazolo Arene Ruthenium Complexes. J. Med. Chem. 2015, 58, 8475–8490. [Google Scholar] [CrossRef] [PubMed]
  22. Grozav, A.; Miclaus, V.; Vostinaru, O.; Ghibu, S.; Berce, C.; Rotar, I.; Mogosan, C.; Therrien, B.; Loghin, F.; Popa, D.-S. Acute toxicity evaluation of a thiazolo arene ruthenium(II) complex in rats. Regul. Toxicol. Pharmacol. 2016, 80, 233–240. [Google Scholar] [CrossRef] [PubMed]
  23. Ignat, A.; Lovasz, T.; Vasilescu, M.; Fischer-fodor, E.; Tatomir, C.B.; Cristea, C.; Silaghi-Dumitrescu, L.; Zaharia, V. Heterocycles 27. Microwave Assisted Synthesis and Antitumour Activity of Novel Phenothiazinyl-Thiazolyl-Hydrazine Derivatives. Arch. Pharm. Chem. Life Sci. 2012, 345, 574–583. [Google Scholar] [CrossRef] [PubMed]
  24. Patra, M.; Joshi, T.; Pierroz, V.; Ingram, K.; Kaiser, M.; Ferrari, S.; Spingler, B.; Keiser, J.; Gasser, G. DMSO-Mediated ligand dissociation: Renaissance for Biological Activity of N-Heterocyclic-[Ru(η6-arene)Cl2] Drug Candidates. Chem. Eur. J. 2013, 19, 14768–14772. [Google Scholar] [CrossRef] [PubMed]
  25. Jensen, L.J.; Kuhn, M.; Stark, M.; Chaffron, S.; Creevey, C.; Muller, J.; Doerks, T.; Julien, P.; Roth, A.; Simonovic, M.; et al. STRING 8—a global view on proteins and their functional interactions in 630 organisms. Nucleic Acids Res. 2009, 37, D412–D416. [Google Scholar] [CrossRef]
  26. GraphPad Prism Version 6.0 for Windows. GraphPad Software: Boston, MA, USA. Available online: www.graphpad.com (accessed on 1 November 2022).
Scheme 1. Synthesis of [(p-cymene)RuLCl]Cl (RuL1RuL4) from [(p-cymene)RuCl2]2 and the phenothiazinyl-hydrazinyl-thiazole ligands (L1L4).
Scheme 1. Synthesis of [(p-cymene)RuLCl]Cl (RuL1RuL4) from [(p-cymene)RuCl2]2 and the phenothiazinyl-hydrazinyl-thiazole ligands (L1L4).
Inorganics 12 00287 sch001
Figure 1. Antiproliferative effects determined from MTT assays after 24 h incubation with RuL1RuL4 on NBC cells (MCF-7, MDA-MS-231, Hs578T) and normal cells (fR2). Log(conc, nM) = Log(concentration of complexes, nM) (mean ± SD, n = 6).
Figure 1. Antiproliferative effects determined from MTT assays after 24 h incubation with RuL1RuL4 on NBC cells (MCF-7, MDA-MS-231, Hs578T) and normal cells (fR2). Log(conc, nM) = Log(concentration of complexes, nM) (mean ± SD, n = 6).
Inorganics 12 00287 g001
Figure 2. (A) Fluorescence microscopy, showing nuclear features after DAPI staining on normal and NBC cell lines after 24 h incubation (40x magnification). Statistical analysis on various cell lines. (B) Normal cell line fR2. (C) Triple negative breast cancer cell line, Hs578T. (D) Double positive breast cancer cell line (data presented as mean ± SD; p * = 0.039 for compound 2, two-side t-test). (E) Triple negative breast cancer cell line, MDA-MB-231 (data presented as mean ± SD; p ** = 0.0047 for RuL1, p *** = 0.0001 for RuL2, two-side t-test).
Figure 2. (A) Fluorescence microscopy, showing nuclear features after DAPI staining on normal and NBC cell lines after 24 h incubation (40x magnification). Statistical analysis on various cell lines. (B) Normal cell line fR2. (C) Triple negative breast cancer cell line, Hs578T. (D) Double positive breast cancer cell line (data presented as mean ± SD; p * = 0.039 for compound 2, two-side t-test). (E) Triple negative breast cancer cell line, MDA-MB-231 (data presented as mean ± SD; p ** = 0.0047 for RuL1, p *** = 0.0001 for RuL2, two-side t-test).
Inorganics 12 00287 g002aInorganics 12 00287 g002b
Figure 3. Expression profile of selected genes in normal and breast cancer cell lines after incubation with RuL1 and RuL2 for 24 h. (AD) The heatmap presents genes for breast cancer cell lines (color bars represent gene expression fold change: red color indicates the increased level and green indicates the decreased expression level in treated cells); (A1,B1,C1,D1) represents the STRING network [25] for the genes with an altered expression level for genes of at least 1.25-fold increase or decrease with a p-value ≤ 0.05 as effect of RuL1 treatment; (A2,B2,C2,D2) represents the STRING network for the genes with an altered expression level considering the same cut-off values effect of the RuL2 treatment.
Figure 3. Expression profile of selected genes in normal and breast cancer cell lines after incubation with RuL1 and RuL2 for 24 h. (AD) The heatmap presents genes for breast cancer cell lines (color bars represent gene expression fold change: red color indicates the increased level and green indicates the decreased expression level in treated cells); (A1,B1,C1,D1) represents the STRING network [25] for the genes with an altered expression level for genes of at least 1.25-fold increase or decrease with a p-value ≤ 0.05 as effect of RuL1 treatment; (A2,B2,C2,D2) represents the STRING network for the genes with an altered expression level considering the same cut-off values effect of the RuL2 treatment.
Inorganics 12 00287 g003
Table 1. IC50 values of RuL1RuL4 determined from the MTT assays.
Table 1. IC50 values of RuL1RuL4 determined from the MTT assays.
Cell Line NameCompoundIC50 [µM]
MDA-MB-231RuL11.496
RuL20.449
RuL314.125
RuL40.229
Hs578TRuL11.186
RuL20.705
RuL30.095
RuL40.228
MCF-7RuL114.514
RuL20.654
RuL30.019
RuL40.247
fR2RuL10.51
RuL20.144
RuL31.26
RuL412.55
Table 2. Gene expression assessments, as an effect of RuL1 and RuL2 treatment on breast cell lines (cut-off value of expression fold change FC ± 1.25 and p-value ≤ 0.05).
Table 2. Gene expression assessments, as an effect of RuL1 and RuL2 treatment on breast cell lines (cut-off value of expression fold change FC ± 1.25 and p-value ≤ 0.05).
Cell LineCompoundGeneFCp-Value
fR2RuL1CASP8−2.360.0032
p53−1.720.0025
TNFSF10−2.210.0043
RuL2BAK1.480.048
CASP8−1.845.12
p53−1.350.0080
MCF-7RuL1BAK4.040.019
FAS3.430.0091
NAIP2.270.0013
CASP84.510.0099
TNF18.510.0036
XIAP2.110.011
BAD2.270.021
RuL2BAK4.040.011
FAS20.280.0043
NAIP3.980.0057
CASP82.40.0069
TNF24.930.0069
p531.560.16
XIAP2.130.0019
FADD1.910.10
BAD7.220.0044
Hs578TRuL1BAK3.090.0044
CASP82.020.019
TNF2.121.77
XIAP1.650.0056
FADD2.410.0091
BAD2.240.027
TNFSF10−5.50.035
RuL2BAK5.798.86
NAIP3.150.022
CASP83.370.0011
TNF12.360.00011
p531.890.0034
NOD12.310.0060
XIAP4.443.61
FADD5.340.0015
BAD2.920.0068
MDA-MB-231RuL1NOD11.440.030
p53−2.090.0021
RuL2p53−1.580.0056
XIAP−1.720.0047
TNFSF10−2.320.014
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

Grozav, A.; Cheminel, T.; Jurj, A.; Zanoaga, O.; Raduly, L.; Braicu, C.; Berindan-Neagoe, I.; Crisan, O.; Gaina, L.; Therrien, B. Arene Ruthenium Complexes Specifically Inducing Apoptosis in Breast Cancer Cells. Inorganics 2024, 12, 287. https://doi.org/10.3390/inorganics12110287

AMA Style

Grozav A, Cheminel T, Jurj A, Zanoaga O, Raduly L, Braicu C, Berindan-Neagoe I, Crisan O, Gaina L, Therrien B. Arene Ruthenium Complexes Specifically Inducing Apoptosis in Breast Cancer Cells. Inorganics. 2024; 12(11):287. https://doi.org/10.3390/inorganics12110287

Chicago/Turabian Style

Grozav, Adriana, Thomas Cheminel, Ancuta Jurj, Oana Zanoaga, Lajos Raduly, Cornelia Braicu, Ioana Berindan-Neagoe, Ovidiu Crisan, Luiza Gaina, and Bruno Therrien. 2024. "Arene Ruthenium Complexes Specifically Inducing Apoptosis in Breast Cancer Cells" Inorganics 12, no. 11: 287. https://doi.org/10.3390/inorganics12110287

APA Style

Grozav, A., Cheminel, T., Jurj, A., Zanoaga, O., Raduly, L., Braicu, C., Berindan-Neagoe, I., Crisan, O., Gaina, L., & Therrien, B. (2024). Arene Ruthenium Complexes Specifically Inducing Apoptosis in Breast Cancer Cells. Inorganics, 12(11), 287. https://doi.org/10.3390/inorganics12110287

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