1
|
Chen L, Tang H, Hu T, Wang J, Ouyang Q, Zhu X, Wang R, Huang W, Huang Z, Chen J. Three Ru(II) complexes modulate the antioxidant transcription factor Nrf2 to overcome cisplatin resistance. J Inorg Biochem 2024; 259:112666. [PMID: 39029397 DOI: 10.1016/j.jinorgbio.2024.112666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/14/2024] [Accepted: 07/12/2024] [Indexed: 07/21/2024]
Abstract
Here, we designed, synthesized and characterized three new cyclometalated Ru(II) complexes, [Ru(phen)2(1-(4-Ph-Ph)-IQ)]+ (phen = 1,10-phenanthroline, IQ = isoquinoline, RuIQ9), [Ru(phen)2(1-(4-Ph-Ph)-7-OCH3-IQ)]+ (RuIQ10), and [Ru(phen)2(1-(4-Ph-Ph)-6,7-(OCH3)2-IQ)]+ (RuIQ11). The cytotoxicity experiments conducted on both 2D and 3D multicellular tumor spheroids (MCTSs) indicated that complexes RuIQ9-11 exhibited notably higher cytotoxicity against A549 and A549/DDP cells when compared to the ligands and precursor compounds as well as clinical cisplatin. Moreover, the Ru(II) complexes displayed low toxicity when tested on normal HBE cells in vitro and exposed to zebrafish embryos in vivo. In addition, complexes RuIQ9-11 could inhibit A549 and A549/DDP cell migration and proliferation by causing cell cycle arrest, mitochondrial dysfunction, and elevating ROS levels to induce apoptosis in these cells. Mechanistic studies revealed that RuIQ9-11 could suppress the expression of Nrf2 and its downstream antioxidant protein HO-1 by inhibiting Nrf2 gene transcription in drug-resistant A549/DDP cells. Simultaneously, they inhibited the expression of efflux proteins MRP1 and p-gp in drug-resistant cells, ensuring the accumulation of the complexes within the cells. This led to an increase in intracellular ROS levels in drug-resistant cells, ultimately causing damage and cell death, thus overcoming cisplatin resistance. More importantly, RuIQ11 could effectively inhibit the migration and proliferation of drug-resistant cells within zebrafish, addressing the issue of cisplatin resistance. Accordingly, the prepared Ru(II) complexes possess significant potential for development as highly effective and low-toxicity lung cancer therapeutic agents to overcome cisplatin resistance.
Collapse
Affiliation(s)
- Lanmei Chen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China
| | - Hong Tang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Tianling Hu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Jie Wang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Qianqian Ouyang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Xufeng Zhu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| | - Rui Wang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Wenyong Huang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Zunnan Huang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China.
| | - Jincan Chen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China.
| |
Collapse
|
2
|
Palmeira-Mello MV, Costa AR, de Oliveira LP, Blacque O, Gasser G, Batista AA. Exploring the potential of ruthenium(II)-phosphine-mercapto complexes as new anticancer agents. Dalton Trans 2024; 53:10947-10960. [PMID: 38895770 DOI: 10.1039/d4dt01191k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The search for new metal-based anticancer drug candidates is a fundamental task in medicinal inorganic chemistry. In this work, we assessed the potential of two new Ru(II)-phosphine-mercapto complexes as potential anticancer agents. The complexes, with the formula [Ru(bipy)(dppen)(Lx)]PF6 [(1), HL1 = 2-mercapto-pyridine and (2), HL2 = 2-mercapto-pyrimidine, bipy = 2,2'-bipyridine, dppen = cis-1,2-bis(diphenylphosphino)-ethylene] were synthesized and characterized by nuclear magnetic resonance (NMR) [1H, 31P(1H), and 13C], high resolution mass spectrometry (HR-MS), cyclic voltammetry, infrared and UV-Vis spectroscopies. Complex 2 was obtained as a mixture of two isomers, 2a and 2b, respectively. The composition of these metal complexes was confirmed by elemental analysis and liquid chromatography-mass spectrometry (LC-MS). To obtain insights into their lipophilicity, their distribution coefficients between n-octanol/PBS were determined. Both complexes showed affinity mainly for the organic phase, presenting positive log P values. Also, their stability was confirmed over 48 h in different media (i.e., DMSO, PBS and cell culture medium) via HPLC, UV-Vis and 31P{1H} NMR spectroscopies. Since enzymes from the P-450 system play a crucial role in cellular detoxification and metabolism, the microsomal stability of 1, which was found to be the most interesting compound of this study, was investigated using human microsomes to verify its potential oxidation in the liver. The analyses by LC-MS and ESI-MS reveal three main metabolites, obtained by oxidation in the dppen and bipy moieties. Moreover, 1 was able to interact with human serum albumin (HSA). The cytotoxicity of the metal complexes was tested in different cancerous and non-cancerous cell lines. Complex 1 was found to be more selective than cisplatin against MDA-MB-231 breast cancer cells when compared to MCF-10A non-cancerous cells. In addition, complex 1 affects cell morphology and migration, and inhibits colony formation in MDA-MB-231 cells, making it a promising cytotoxic agent against breast cancer.
Collapse
Affiliation(s)
- Marcos V Palmeira-Mello
- Departament of Chemistry, Universidade Federal de São Carlos, 13561-901, São Carlos, SP, Brazil.
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Paris, France.
| | - Analu R Costa
- Departament of Chemistry, Universidade Federal de São Carlos, 13561-901, São Carlos, SP, Brazil.
| | - Leticia P de Oliveira
- Departament of Chemistry, Universidade Federal de São Carlos, 13561-901, São Carlos, SP, Brazil.
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Paris, France.
| | - Alzir A Batista
- Departament of Chemistry, Universidade Federal de São Carlos, 13561-901, São Carlos, SP, Brazil.
| |
Collapse
|
3
|
Abirami A, Devan U, Ramesh R, Antony Joseph Velanganni A, Małecki JG. Exploring the cytotoxicity of dinuclear Ru(II) p-cymene complexes appended N, N'-bis(4-substituted benzoyl)hydrazines: insights into the mechanism of apoptotic cell death. Dalton Trans 2024; 53:5167-5179. [PMID: 38380977 DOI: 10.1039/d3dt04234k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Cancer is a perilous life-threatening disease, and attempts are constantly being made to create multinuclear transition metal complexes that could lead to the development of potential anticancer medications and administration procedures. Hence, this work aims to design, synthesize, characterize, and assess the anticancer efficacy of ruthenium p-cymene complexes incorporating N,N'-bis(4-substituted benzoyl)hydrazine ligands. The formation of the new complexes (Ru2H1-Ru2H3) has been thoroughly established by elemental analysis, and FT-IR, UV-vis, NMR, and HR-MS spectral techniques. The solid-state molecular structures of the complexes Ru2H1 and Ru2H3 have been determined using the SC-XRD study, which confirms the N, O, and Cl-legged piano stool pseudo-octahedral geometry of each ruthenium(II) ion. The stability of these complexes in the solution state and their lipophilicity profile have been determined. Furthermore, the title complexes were tested for their in vitro anticancer activity against cancerous H460 (lung cancer cells), SkBr3 (breast cancer cells), HepG2 (liver cancer cells), and HeLa (cervical cancer cells) along with non-cancerous (HEK-293) cells. The IC50 results revealed that complex Ru2H3 exhibits potent activity against the proliferation of all four cancer cells and outscored the effect of the standard metallodrug cisplatin. This may be attributed to the presence of a couple of lipophilic electron-donating methoxy groups in the ligand scaffold and also the ruthenium(II) p-cymene motifs. Advantageously, all the complexes (Ru2H1-Ru2H3) displayed cytotoxic specificity only towards cancerous cells by leaving the off-target non-cancerous cells undamaged. Acridine orange/ethidium bromide (AO/EB) staining, Hoechst 33342, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) staining assays were used to investigate the apoptotic pathway and ROS levels in mitochondria. The results of western blot analysis confirmed that the complexes triggered apoptosis through an intrinsic mitochondrial pathway by upregulating Bax and downregulating Bcl-2 proteins. Finally, the extent of apoptosis triggered by the complex Ru2H3 was quantified with the aid of flow cytometry using the Annexin V-FITC/propidium iodide (PI) double-staining technique.
Collapse
Affiliation(s)
- Arunachalam Abirami
- Centre for Organometallic Chemistry, School of Chemistry, Bharathidasan University, Tiruchirappalli - 620 024, India.
| | - Umapathy Devan
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli - 620 024, India
| | - Rengan Ramesh
- Centre for Organometallic Chemistry, School of Chemistry, Bharathidasan University, Tiruchirappalli - 620 024, India.
| | - Arockiam Antony Joseph Velanganni
- Molecular Oncology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli - 620 024, India
| | - Jan Grzegorz Małecki
- Department of Crystallography, Institute of Chemistry, University of Silesia, Katowice, Poland
| |
Collapse
|
4
|
de Melo MRS, Ribeiro AB, Fernandes G, Squarisi IS, de Melo Junqueira M, Batista AA, da Silva MM, Tavares DC. Ruthenium(II) complex with 2-mercaptothiazoline ligand induces selective cytotoxicity involving DNA damage and apoptosis in melanoma cells. J Biol Inorg Chem 2024; 29:159-168. [PMID: 38182820 DOI: 10.1007/s00775-023-02036-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/26/2023] [Indexed: 01/07/2024]
Abstract
Melanoma is the most aggressive and lethal type of skin cancer due to its characteristics such as high metastatic potential and low response rate to existing treatment modalities. In this way, new drug prototypes are being studied to solve the problem of treating patients with melanoma. Among these, ruthenium-based metallopharmaceuticals may be promising alternatives due to their antitumor characteristics and low systemic toxicity. In this context, the present study evaluated the antineoplastic effect of the ruthenium complex [Ru(mtz)(dppe)2]PF6-2-mercaptothiazoline-di-1,2-bis(diphenylphosphine) ethaneruthenium(II), namely RuMTZ, on human melanoma (A-375) and murine (B16-F10) cells, considering different approaches. Through XTT colorimetric and clonogenic efficiency assays, the complex revealed the selective cytotoxic activity, with the lowest IC50 (0.4 µM) observed for A375 cells. RuMTZ also induced changes in cell morphology, increased cell population in the sub-G0 phase and inhibiting cell migration. The levels of γH2AX and cleaved caspase 3 proteins were increased in both cell lines treated with RuMTZ. These findings indicated that the cytotoxic activity of RuMTZ on melanoma cells is related, at least in part, to the induction of DNA damage and apoptosis. Therefore, RuMTZ exhibited promising antineoplastic activity against melanoma cells.
Collapse
Affiliation(s)
| | | | - Gabriela Fernandes
- Laboratory of Mutagenesis, University of Franca, Franca, São Paulo, 14404-600, Brazil
| | - Iara Silva Squarisi
- Laboratory of Mutagenesis, University of Franca, Franca, São Paulo, 14404-600, Brazil
| | | | - Alzir Azevedo Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13565-905, Brazil
| | - Monize Martins da Silva
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13565-905, Brazil
| | | |
Collapse
|
5
|
Grawe GF, Oliveira KM, Leite CM, de Oliveira TD, Costa AR, Moraes CA, Honorato J, Cominetti MR, Castellano EE, Correa RS, Machado SP, Batista AA. Cytotoxic activity of Ru(II)/DPEPhos/N,S-mercapto complexes (DPEPhos -[(2-diphenylphosphino)phenyl]ether). J Inorg Biochem 2023; 244:112204. [PMID: 37004320 DOI: 10.1016/j.jinorgbio.2023.112204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/15/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
We report here on three new ruthenium(II) complexes, [Ru(DPEPhos)(mtz)(bipy)]PF6 (Ru1), [Ru(DPEPhos)(mmi)(bipy)]PF6 (Ru2) and [Ru(DPEPhos)(dmp)(bipy)]PF6 (Ru3). DPEPhos = bis-[(2-diphenylphosphino)phenyl]ether, mtz = 2-mercapto-2-thiazoline, mmi = 2-mercapto-1-methylimidazole, dmp = 4,6-diamino-2-mercaptopyrimidine and bipy = 2,2'-bipyridine. The compounds were characterized by several spectroscopic techniques, and the molecular structure of Ru1 complex was determined by single-crystal X-ray diffraction. The cytotoxicity of Ru1 - Ru3 complexes were tested against the A549 (human lung) and the MDA-MB-231 (human breast) cancer cell lines and against MRC-5 (non-tumor lung) and MCF-10A (non-tumor breast) cell lines through the MTT assay. All three complexes are cytotoxic against the cell lines studied, with IC50 values lower than those found for the cisplatin. Among them, the Ru2 complex has shown the best selectivity against MDA-MB-231 cancer cell lines, with an IC50 value 12 times lower than that on MCF-10A. The complex Ru2 was capable to induce changes in MDA-MB-231 cells morphology, with loss of cellular adhesion, inhibited colony formation and induce an accumulation of cells at the sub-G1 phase, with an increase in S-phase and decrease of cells at G2 phase. Viscosity, electrochemical and Hoechst 33258 displacement experiments for Ru1 - Ru3 complexes with calf thymus DNA (CT-DNA) showed an electrostatic and groove binding mode of interaction. Additionally, the complexes interact with the protein Human Serum Albumin (HSA) by static mechanism. The negative values for ΔH and ΔS indicate that van der Waals forces and hydrogen bonding may occurs between the complexes and HSA. Therefore, this class of complexes are promising anticancer candidates and may be selected to further detailed studies.
Collapse
|
6
|
Gonçalves YG, Becceneri AB, Graminha AE, Miranda VM, Rios RR, Rinaldi-Neto F, Costa MS, Gonçalves ACR, Deflon VM, Yoneyama KAG, Maia PIS, Franca EF, Cominetti MR, Silva RS, Von Poelhsitz G. New ruthenium(II) complexes with cyclic thio- and semicarbazone: evaluation of cytotoxicity and effects on cell migration and apoptosis of lung cancer cells. Dalton Trans 2023. [PMID: 37377063 DOI: 10.1039/d3dt00750b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
We describe the synthesis, physicochemical characterization, and in vitro antitumor assays of four novel analogous ruthenium(II) complexes with general formula cis-[RuII(N-L)(P-P)2]PF6, where P-P = bis(diphenylphosphine)methane (dppm, in complexes 1 and 2) or bis(diphenylphosphine)ethane (dppe, in complexes 3 and 4) and N-L = 5,6-diphenyl-4,5-dihydro-2H-[1,2,4]triazine-3-thione (Btsc, in complexes 1 and 3) or 5,6-diphenyltriazine-3-one (Bsc, in complexes 2 and 4). The data were consistent with cis arrangement of the biphosphine ligands. For the Btsc and Bsc ligands, the data pointed to monoanionic bidentate coordination to ruthenium(II) through N,S and N,O, respectively. Single-crystal X-ray diffraction showed that complex 1 crystallized in the monoclinic system, space group P21/c. Determination of the cytotoxicity profiles of complexes 1-4 gave SI values ranging from 1.19 to 3.50 against the human lung adenocarcinoma cell line A549 and the non-tumor lung cell line MRC-5. Although the molecular docking studies suggested that the interaction between DNA and complex 4 was energetically favorable, the experimental results showed that they interacted weakly. Overall, our results demonstrated that these novel ruthenium(II) complexes have interesting in vitro antitumor potential and this study may contribute to further studies in medicinal inorganic chemistry.
Collapse
Affiliation(s)
- Yasmim G Gonçalves
- Chemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil.
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Amanda B Becceneri
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Angelica E Graminha
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
- Gerontology Department, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Victor M Miranda
- Grupo de Química Inorgânica Estrutural e Biológica, Chemistry Institute of São Carlos, Universidade de São Paulo, USP - São Carlos, São Carlos, SP, Brazil
| | - Rafaella R Rios
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Francisco Rinaldi-Neto
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mônica S Costa
- Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Ana C R Gonçalves
- Exacts, Natural Sciences, and Education Institute, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Victor M Deflon
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kelly A G Yoneyama
- Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Pedro I S Maia
- Exacts, Natural Sciences, and Education Institute, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Eduardo F Franca
- Laboratório de Cristalografia e Química Computacional, Chemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Márcia R Cominetti
- Gerontology Department, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Roberto S Silva
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | |
Collapse
|
7
|
Yusoh NA, Tiley PR, James SD, Harun SN, Thomas JA, Saad N, Hii LW, Chia SL, Gill MR, Ahmad H. Discovery of Ruthenium(II) Metallocompound and Olaparib Synergy for Cancer Combination Therapy. J Med Chem 2023; 66:6922-6937. [PMID: 37185020 DOI: 10.1021/acs.jmedchem.3c00322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Synergistic drug combinations can extend the use of poly(ADP-ribose) polymerase inhibitors (PARPi) such as Olaparib to BRCA-proficient tumors and overcome acquired or de novo drug resistance. To identify new synergistic combinations for PARPi, we screened a "micro-library" comprising a mix of commercially available drugs and DNA-binding ruthenium(II) polypyridyl complexes (RPCs) for Olaparib synergy in BRCA-proficient triple-negative breast cancer cells. This identified three hits: the natural product Curcumin and two ruthenium(II)-rhenium(I) polypyridyl metallomacrocycles. All combinations identified were effective in BRCA-proficient breast cancer cells, including an Olaparib-resistant cell line, and spheroid models. Mechanistic studies indicated that synergy was achieved via DNA-damage enhancement and resultant apoptosis. Combinations showed low cytotoxicity toward non-malignant breast epithelial cells and low acute and developmental toxicity in zebrafish embryos. This work identifies RPC metallomacrocycles as a novel class of agents for cancer combination therapy and provides a proof of concept for the inclusion of metallocompounds within drug synergy screens.
Collapse
Affiliation(s)
- Nur Aininie Yusoh
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor, Malaysia
| | - Paul R Tiley
- Department of Chemistry, Faculty of Science and Engineering, Swansea University, Swansea SA2 8PP, U.K
| | - Steffan D James
- Department of Chemistry, Faculty of Science and Engineering, Swansea University, Swansea SA2 8PP, U.K
| | - Siti Norain Harun
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor, Malaysia
| | - Jim A Thomas
- Department of Chemistry, University of Sheffield, Sheffield S3 7HF, U.K
| | - Norazalina Saad
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor, Malaysia
| | - Ling-Wei Hii
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Suet Lin Chia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor, Malaysia
| | - Martin R Gill
- Department of Chemistry, Faculty of Science and Engineering, Swansea University, Swansea SA2 8PP, U.K
| | - Haslina Ahmad
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
8
|
de Araujo-Neto JH, Guedes APM, Leite CM, Moraes CAF, Santos AL, Brito RDS, Rocha TL, Mello-Andrade F, Ellena J, Batista AA. "Half-Sandwich" Ruthenium Complexes with Alizarin as Anticancer Agents: In Vitro and In Vivo Studies. Inorg Chem 2023; 62:6955-6969. [PMID: 37099760 DOI: 10.1021/acs.inorgchem.3c00183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Upon exploration of the chemistry of the combination of ruthenium/arene with anthraquinone alizarin (L), three new complexes with the general formulas [Ru(L)Cl(η6-p-cymene)] (C1), [Ru(L)(η6-p-cymene)(PPh3)]PF6 (C2), and [Ru(L)(η6-p-cymene)(PEt3)]PF6 (C3) were synthesized and characterized using spectroscopic techniques (mass, IR, and 1D and 2D NMR), molar conductivity, elemental analysis, and X-ray diffraction. Complex C1 exhibited fluorescence, such as free alizarin, while in C2 and C3, the emission was probably quenched by monophosphines and the crystallographic data showed that hydrophobic interactions are predominant in intermolecular contacts. The cytotoxicity of the complexes was evaluated in the MDA-MB-231 (triple-negative breast cancer), MCF-7 (breast cancer), and A549 (lung) tumor cell lines and MCF-10A (breast) and MRC-5 (lung) nontumor cell lines. Complexes C1 and C2 were more selective to the breast tumor cell lines, and C2 was the most cytotoxic (IC50 = 6.5 μM for MDA-MB-231). In addition, compound C1 performs a covalent interaction with DNA, while C2 and C3 present only weak interactions; however, internalization studies by flow cytometry and confocal microscopy showed that complex C1 does not accumulate in viable MDA-MB-231 cells and is detected in the cytoplasm only after cell permeabilization. Investigations of the mechanism of action of the complexes indicate that C2 promotes cell cycle arrest in the Sub-G1 phase in MDA-MB-231, inhibits its colony formation, and has a possible antimetastatic action, impeding cell migration in the wound-healing experiment (13% of wound healing in 24 h). The in vivo toxicological experiments with zebrafish indicate that C1 and C3 exhibit the most zebrafish embryo developmental toxicity (inhibition of spontaneous movements and heartbeats), while C2, the most promising anticancer drug in the in vitro preclinical tests, revealed the lowest toxicity in in vivo preclinical screening.
Collapse
Affiliation(s)
- João Honorato de Araujo-Neto
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Adriana P M Guedes
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Celisnolia M Leite
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
| | - Carlos André F Moraes
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Andressa L Santos
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Rafaella da S Brito
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Thiago L Rocha
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Francyelli Mello-Andrade
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
- Instituto Federal de Educação Ciência e Tecnologia (IFG), Goiânia, Goiás 74055-110, Brazil
| | - Javier Ellena
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| |
Collapse
|
9
|
Leitao RCF, Silva F, Ribeiro GH, Santos IC, Guerreiro JF, Mendes F, Batista AA, Pavan FR, da S Maia PI, Paulo A, Deflon VM. Gallium and indium complexes with isoniazid-derived ligands: Interaction with biomolecules and biological activity against cancer cells and Mycobacterium tuberculosis. J Inorg Biochem 2023; 240:112091. [PMID: 36527994 DOI: 10.1016/j.jinorgbio.2022.112091] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Gallium and indium octahedral complexes with isoniazid derivative ligands were successfully prepared. The ligands, isonicotinoyl benzoylacetone (H2L1) and 4-chlorobenzoylacetone isonicotinoyl hydrazone (H2L2), and their respective coordination compounds with gallium and indium [GaL1(HL1)] (GaL1), [GaL2(HL2)] (GaL2), [InL1(HL1)] (InL1) and [InL2(HL2)] (InL2) were investigated by NMR, ESI-MS, UV-Vis, IR, single-crystal X-ray diffraction and elemental analysis. In vitro interaction studies with human serum albumin (HSA) evidenced a moderate affinity of all complexes with HSA through spontaneous hydrophobic interactions. The greatest suppression of HSA fluorescence was caused by GaL2 and InL2, which was associated to the higher lipophilicity of H2L2. In vitro interaction studies with CT-DNA indicated weak interactions of the biomolecule with all complexes. Cytotoxicity assays with MCF-7 (breast carcinoma), PC-3 (prostate carcinoma) and RWPE-1 (healthy human prostate epithelial) cell lines showed that complexes with H2L2 are more active and selective against MCF-7, with the greatest cytotoxicity observed for InL2 (IC50 = 10.34 ± 1.69 μM). H2L1 and H2L2 were labelled with gallium-67, and it was verified that 67GaL2 has a greater lipophilicity than 67GaL1, as well as higher stability in human serum or in the presence of apo-transferrin. Cellular uptake assays with 67GaL1 and 67GaL2 evidenced that the H2L2-containing radiocomplex has a higher accumulation in MCF-7 and PC-3 cells than the non-halogenated congener 67GaL1. The anti-Mycobacterium tuberculosis assays revealed that both ligands and metal complexes are potent growth inhibitors, with MIC90 (μg mL-1) values observed from 0.419 ± 0.05 to 1.378 ± 0.21.
Collapse
Affiliation(s)
- Renan C F Leitao
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil
| | - Francisco Silva
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Gabriel H Ribeiro
- Departamento de Química, Universidade Federal de São Carlos, 13565-905 São Carlos, SP, Brazil
| | - Isabel C Santos
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal; Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Joana F Guerreiro
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Filipa Mendes
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal; Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, 13565-905 São Carlos, SP, Brazil
| | - Fernando R Pavan
- Faculdade de Ciências Farmacêuticas, UNESP - Universidade Estadual Paulista, Campus Araraquara, 14.800-903 Araraquara, SP, Brazil
| | - Pedro Ivo da S Maia
- Departamento de Química, Universidade Federal do Triângulo Mineiro, 38025-440 Uberaba, MG, Brazil
| | - António Paulo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal; Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Victor M Deflon
- Instituto de Química de São Carlos, Universidade de São Paulo, 13566-590 São Carlos, SP, Brazil.
| |
Collapse
|
10
|
Synergy of ruthenium metallo-intercalator, [Ru(dppz) 2(PIP)] 2+, with PARP inhibitor Olaparib in non-small cell lung cancer cells. Sci Rep 2023; 13:1456. [PMID: 36702871 PMCID: PMC9879939 DOI: 10.1038/s41598-023-28454-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) are critical DNA repair enzymes that are activated as part of the DNA damage response (DDR). Although inhibitors of PARP (PARPi) have emerged as small molecule drugs and have shown promising therapeutic effects, PARPi used as single agents are clinically limited to patients with mutations in germline breast cancer susceptibility gene (BRCA). Thus, novel PARPi combination strategies may expand their usage and combat drug resistance. In recent years, ruthenium polypyridyl complexes (RPCs) have emerged as promising anti-cancer candidates due to their attractive DNA binding properties and distinct mechanisms of action. Previously, we reported the rational combination of the RPC DNA replication inhibitor [Ru(dppz)2(PIP)]2+ (dppz = dipyrido[3,2-a:2',3'-c]phenazine, PIP = 2-(phenyl)-imidazo[4,5-f][1,10]phenanthroline), "Ru-PIP", with the PARPi Olaparib in breast cancer cells. Here, we expand upon this work and examine the combination of Ru-PIP with Olaparib for synergy in lung cancer cells, including in 3D lung cancer spheroids, to further elucidate mechanisms of synergy and additionally assess toxicity in a zebrafish embryo model. Compared to single agents alone, Ru-PIP and Olaparib synergy was observed in both A549 and H1975 lung cancer cell lines with mild impact on normal lung fibroblast MRC5 cells. Employing the A549 cell line, synergy was confirmed by loss in clonogenic potential and reduced migration properties. Mechanistic studies indicated that synergy is accompanied by increased double-strand break (DSB) DNA damage and reactive oxygen species (ROS) levels which subsequently lead to cell death via apoptosis. Moreover, the identified combination was successfully able to inhibit the growth of A549 lung cancer spheroids and acute zebrafish embryos toxicity studies revealed that this combination showed reduced toxicity compared to single-agent Ru-PIP.
Collapse
|
11
|
Hu X, Guo L, Liu M, Zhang Q, Gong Y, Sun M, Feng S, Xu Y, Liu Y, Liu Z. Increasing Anticancer Activity with Phosphine Ligation in Zwitterionic Half-Sandwich Iridium(III), Rhodium(III), and Ruthenium(II) Complexes. Inorg Chem 2022; 61:20008-20025. [PMID: 36426422 DOI: 10.1021/acs.inorgchem.2c03279] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The synthesis and biological assessment of neutral or cationic platinum group metal-based anticancer complexes have been extremely studied, whereas there are few reports on the corresponding zwitterionic complexes. Herein, the synthesis, characterization, and bioactivity of zwitterionic half-sandwich phosphine-imine iridium(III), rhodium(III), and ruthenium(II) complexes were presented. The sulfonated phosphine-imine ligand and a group of zwitterionic half-sandwich P,N-chelating organometallic complexes were fully characterized by nuclear magnetic resonance (NMR), mass spectrum (electrospray ionization, ESI), elemental analysis, and X-ray crystallography. The solution stability of these complexes and their spectral properties were also determined. Notably, almost all of these complexes showed enhanced anticancer activity against model HeLa and A549 cancer cells than the corresponding zwitterionic pyridyl-imine N,N-chelating iridium(III) and ruthenium(II) complexes, which have exhibited inactive or low active in our previous work. The increase in the lipophilic property and intracellular uptake levels of these zwitterionic P,N-chelating complexes appeared to be associated with their superior cytotoxicity. In addition, these complexes showed biomolecular interactions with bovine serum albumin (BSA). The flow cytometry studies indicated that the representative complex Ir1 could induce early-stage apoptosis in A549 cells. Further, confocal microscopy imaging analysis displayed that Ir1 entered A549 cells through the energy-dependent pathway, targeted lysosome, and could cause lysosomal damage. In particular, these complexes could impede cell migration in A549 cells.
Collapse
Affiliation(s)
- Xueyan Hu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Lihua Guo
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Mengqi Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Qiuya Zhang
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yuwen Gong
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Mengru Sun
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Shenghan Feng
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Youzhi Xu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yiming Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Zhe Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| |
Collapse
|
12
|
Abubakar A, Abdulmalek E, Norhamidah Wan Ibrahim W, Cordova KE, Abdul Rahman MB. ZIF-90 nanoparticles modified with a homing peptide for targeted delivery of cisplatin. Front Chem 2022; 10:1076350. [PMID: 36545218 PMCID: PMC9760700 DOI: 10.3389/fchem.2022.1076350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/21/2022] [Indexed: 12/11/2022] Open
Abstract
To improve the selective delivery of cisplatin (Cis) to cancer cells, we report and establish the significance of active, targeting drug delivery nanosystems for efficient treatment of lung cancer. Specifically, pH-responsive nano-sized zeolitic imidazolate framework (nZIF-90) was synthesized, post-synthetically modified with an Arg-Gly-Asp peptide motif (RGD@nZIF-90), a known cancer cell homing peptide, and loaded with a large amount of Cis (RGD@Cis⊂nZIF-90). RGD@Cis⊂nZIF-90 was shown to be highly stable under physiological conditions (pH = 7.4) with framework dissociation occurring under slightly acidic conditions (pH = 5.0)-conditions relevant to tumor cells-from which 90% of the encapsulated Cis was released in a sustained manner. In vitro assays demonstrated that RGD@Cis⊂nZIF-90 achieved significantly better cytotoxicity (65% at 6.25 μg ml-1) and selectivity (selectivity index = 4.18 after 48 h of treatment) against adenocarcinoma alveolar epithelial cancer cells (A549) when compared with the unmodified Cis⊂nZIF-90 (22%). Cellular uptake using A549 cells indicated that RGD@Cis⊂nZIF-90 was rapidly internalized leading to significant cell death. After successfully realizing this nanocarrier system, we demonstrated its efficacy in transporting and delivering Cis to cancer cells.
Collapse
Affiliation(s)
- Adamu Abubakar
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Seri Kembangan, Malaysia,Department of Chemical Sciences, Taraba State University, P.M.B, Taraba State, Jalingo, Nigeria
| | - Emilia Abdulmalek
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Seri Kembangan, Malaysia,*Correspondence: Emilia Abdulmalek, ; Kyle E. Cordova,
| | | | - Kyle E. Cordova
- Materials Discovery Research Unit, Advanced Research Centre, Royal Scientific Society, Amman, Jordan,Department of Chemistry, Faculty of Science, UPM, Selangor Darul Ehsan, Malaysia,Foundry of Reticular Materials for Sustainability (FORMS), Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology, UPM, Selangor Darul Ehsan, Malaysia,*Correspondence: Emilia Abdulmalek, ; Kyle E. Cordova,
| | - Mohd Basyaruddin Abdul Rahman
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Seri Kembangan, Malaysia,Department of Chemistry, Faculty of Science, UPM, Selangor Darul Ehsan, Malaysia,Foundry of Reticular Materials for Sustainability (FORMS), Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology, UPM, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
13
|
de Oliveira TD, Ribeiro GH, Honorato J, Leite CM, Santos ACDS, Silva ED, Pereira VRA, Plutín AM, Cominetti MR, Castellano EE, Batista AA. Cytotoxic and antiparasitic activities of diphosphine-metal complexes of group 10 containing acylthiourea as ligands. J Inorg Biochem 2022; 234:111906. [PMID: 35759891 DOI: 10.1016/j.jinorgbio.2022.111906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022]
Abstract
In this work, group 10 transition metal complexes bearing dppe [1,2-bis(diphenylphosphino)ethane] and acylthiourea ligands were evaluated for their cytotoxic and antiparasitic activities. Six new complexes with a general formula [M(Ln)(dppe)]BF4 [where M = NiII, PdII or PtII; Ln = N, N'-dimethyl-N-benzoyl thiourea (L1) or N, N'-dimethyl-N-tiofenyl thiourea (L2) were synthesized and characterized by infrared, NMR (31P{1H}, 1H and 13C{1H}) spectroscopies, elemental analysis and molar conductivity. The structures of the complexes were confirmed by X-ray diffraction technique. The biological activity of the complexes was evaluated on breast cancer cells (MDA-MB-231 and MCF-7) and causative agents of chagas disease and leishmaniasis. The complexes presented higher cytotoxicity for breast cancer cell lines compared to non-tumor cells. Nickel complexes stood out when evaluated against the triple-negative breast cancer line (MDA-MB-231), presenting considerably lower IC50 values (about 10 to 22×), when compared to palladium and platinum complexes, and the cisplatin drug. When evaluated on the triple-negative line (MDA-MB-231), the complexes [Ni(L2)(dppe)]BF4(2), [Pd(L2)(dppe)]BF4(4) and [Pt(L2)(dppe)]BF4(6) were able to induce cell morphological changes, influence on the cell colony formation and the size of the cells. The complexes inhibit cell migration and cause changes to the cell cytoskeleton and nuclear arrangement. In the same cell line, the compounds caused cell arrest in the Sub-G1 phase of the cell cycle. The compounds were also tested against the Trypanosom Cruzi (T. cruzi) and Leishmania sp. parasites, which cause Chagas and leishmaniasis disease, respectively. The compounds showed good anti-parasitic activity, mainly for T. cruzi, with lower IC50 values, when compared to the commercial drug, benznidazole. The compounds interact with CT-DNA, indicating that interaction occurs by the minor groove of the biomolecule.
Collapse
Affiliation(s)
- Tamires D de Oliveira
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, 3561-901 São Carlos, SP, Brazil.
| | - Gabriel H Ribeiro
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, 3561-901 São Carlos, SP, Brazil
| | - João Honorato
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, 3561-901 São Carlos, SP, Brazil
| | - Celisnolia M Leite
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, 3561-901 São Carlos, SP, Brazil
| | - Aline Caroline da S Santos
- Fundação Oswaldo Cruz (Fiocruz-Pernambuco), Instituto Aggeu Magalhães, 50670-420 Recife, Pernambuco, Brazil
| | - Elis D Silva
- Fundação Oswaldo Cruz (Fiocruz-Pernambuco), Instituto Aggeu Magalhães, 50670-420 Recife, Pernambuco, Brazil
| | - Valéria Rêgo A Pereira
- Fundação Oswaldo Cruz (Fiocruz-Pernambuco), Instituto Aggeu Magalhães, 50670-420 Recife, Pernambuco, Brazil
| | - Ana M Plutín
- Laboratório de Síntesis Orgánica, Facultad de Química, Universidad de La Habana - UH, 10400 Habana, Cuba
| | - Márcia R Cominetti
- Departamento de Gerontologia, Universidade Federal de São Carlos - UFSCar, 3561-901 São Carlos, SP, Brazil
| | - Eduardo E Castellano
- Instituto de Física de São Carlos, Universidade de São Paulo - USP, 13560-970 São Carlos, SP, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, 3561-901 São Carlos, SP, Brazil.
| |
Collapse
|
14
|
Li S, Jiang C, Tan J, Zhou Q, Yin J, He Y. Sodium arsenite-mediated upregulation of circDHX34 promotes apoptosis in hormone-independent breast cancer cells by regulating apoptotic genes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:2728-2736. [PMID: 34378131 DOI: 10.1007/s11356-021-15891-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Arsenic and the compounds thereof can be carcinogens or therapeutic agents for different cancer types. However, for breast cancer (BC), studies have yielded conflicted results on the role of arsenic. A previous study by the present authors indicated a potential relationship between circDHX34 and sodium arsenite-treated BC cells. As such, the expression, function, and potential mechanism of circDHX34 in sodium arsenite-treated MDA-MB-231 cells were further detected. In the present study, findings were made that sodium arsenite upregulated circDHX34 expression in MDA-MB-231 cells in a dose-dependent manner, and knockdown of circDHX34 could promote cell proliferation and inhibit apoptosis. Further investigations revealed that knockdown of circDHX34 upregulated the expression levels of antiapoptotic genes BCL2 and BCL2L1 and downregulated the expression levels of proapoptotic genes CASP8 and CASP9. To conclude, by regulating apoptotic genes, sodium arsenite-mediated upregulation of circDHX34 promotes apoptosis in hormone-independent breast cancer cells.
Collapse
Affiliation(s)
- Shuting Li
- School of Public Health, Kunming Medical University, No.1168 Chunrong West Road, Chenggong District, Kunming, 650500, Yunnan, China
| | - Chenglan Jiang
- School of Public Health, Kunming Medical University, No.1168 Chunrong West Road, Chenggong District, Kunming, 650500, Yunnan, China
| | - Jingwen Tan
- School of Public Health, Kunming Medical University, No.1168 Chunrong West Road, Chenggong District, Kunming, 650500, Yunnan, China
| | - Qian Zhou
- School of Public Health, Kunming Medical University, No.1168 Chunrong West Road, Chenggong District, Kunming, 650500, Yunnan, China
| | - Jinyao Yin
- School of Public Health, Kunming Medical University, No.1168 Chunrong West Road, Chenggong District, Kunming, 650500, Yunnan, China
| | - Yuefeng He
- School of Public Health, Kunming Medical University, No.1168 Chunrong West Road, Chenggong District, Kunming, 650500, Yunnan, China.
| |
Collapse
|
15
|
Pan NL, Liao JX, Huang MY, Zhang YQ, Chen JX, Zhang ZW, Yang ZX, Long XE, Wu XT, Sun J. Lysosome-targeted ruthenium(II) complexes induce both apoptosis and autophagy in HeLa cells. J Inorg Biochem 2022; 229:111729. [DOI: 10.1016/j.jinorgbio.2022.111729] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022]
|
16
|
|
17
|
Mello-Andrade F, Guedes APM, Pires WC, Velozo-Sá VS, Delmond KA, Mendes D, Molina MS, Matuda L, de Sousa MAM, Melo-Reis P, Gomes CC, Castro CH, Almeida MAP, Menck CFM, Batista AA, Burikhanov R, Rangnekar VM, Silveira-Lacerda E. Ru(II)/amino acid complexes inhibit the progression of breast cancer cells through multiple mechanism-induced apoptosis. J Inorg Biochem 2021; 226:111625. [PMID: 34655962 DOI: 10.1016/j.jinorgbio.2021.111625] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/20/2022]
Abstract
For some cancer subtypes, such as triple-negative breast cancer, there are no specific therapies, which leads to a poor prognosis associated with invasion and metastases. Ruthenium complexes have been developed to act in all steps of tumor growth and its progression. In this study, we investigated the effects of Ruthenium (II) complexes coupled to the amino acids methionine (RuMet) and tryptophan (RuTrp) on the induction of cell death, clonogenic survival ability, inhibition of angiogenesis, and migration of MDA-MB-231 cells (human triple-negative breast cancer). The study also demonstrated that the RuMet and RuTrp complexes induce cell cycle blockage and apoptosis of MDA-MB-231 cells, as evidenced by an increase in the number of Annexin V-positive cells, p53 phosphorylation, caspase 3 activation, and poly(ADP-ribose) polymerase cleavage. Moreover, morphological changes and loss of mitochondrial membrane potential were detected. The RuMet and RuTrp complexes induced DNA damage probably due to reactive oxygen species production related to mitochondrial membrane depolarization. Therefore, the RuMet and RuTrp complexes acted directly on breast tumor cells, leading to cell death and inhibiting their metastatic potential; this reveals the potential therapeutic action of these drugs.
Collapse
Affiliation(s)
- Francyelli Mello-Andrade
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil; Department of Chemistry, Federal Institute of Education, Science and Technology of Goiás, Goiânia, Goiás 74055-110, Brazil.
| | - Adriana P M Guedes
- Department of Chemistry, Federal University of São Carlos, São Carlos, SP 13565-905, Brazil
| | - Wanessa C Pires
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil
| | - Vivianne S Velozo-Sá
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil
| | - Kezia A Delmond
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil
| | - Davi Mendes
- Department of Microbiology, Laboratory of DNA Repair, Institute of Biomedical Sciences, University of São Paulo - USP, São Paulo, SP 05508-900, Brazil
| | - Matheus S Molina
- Department of Microbiology, Laboratory of DNA Repair, Institute of Biomedical Sciences, University of São Paulo - USP, São Paulo, SP 05508-900, Brazil
| | - Larissa Matuda
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás - UFG, Goiânia, GO 74690-900, Brazil
| | | | - Paulo Melo-Reis
- Departament of Biomedicine, Pontifical Catholic University of Goiás, Goiânia, GO, Brazil
| | - Clever C Gomes
- Department of Morphology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO 74690-900, Brazil
| | - Carlos Henrique Castro
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás - UFG, Goiânia, GO 74690-900, Brazil
| | - Márcio Aurélio P Almeida
- Coordination of Science and Technology, Federal University of Maranhão, São Luís, MA 65080-805, Brazil
| | - Carlos F M Menck
- Department of Microbiology, Laboratory of DNA Repair, Institute of Biomedical Sciences, University of São Paulo - USP, São Paulo, SP 05508-900, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, SP 13565-905, Brazil
| | - Ravshan Burikhanov
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, United States of America
| | - Vivek M Rangnekar
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, United States of America; L. P. Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States of America
| | - Elisângela Silveira-Lacerda
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil.
| |
Collapse
|
18
|
Chen BC, Lu JJ, Jiang N, Ma XR, Li RT, Ye RR. Synthesis, characterization and antitumor mechanism investigation of ruthenium(II) polypyridyl complexes with artesunate moiety. J Biol Inorg Chem 2021; 26:909-918. [PMID: 34545414 DOI: 10.1007/s00775-021-01901-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022]
Abstract
Six artesunate (ART) conjugated ruthenium(II) complexes (Ru(II)-ART conjugates) with the formula [Ru(N^N)2bpy(4-CH3-4'-CH2OART)](PF6)2 (Ru-ART-1-3) and [Ru(N^N)2bpy(4-CH2OART-4'-CH2OART)](PF6)2 (Ru-ART-4-6) (N^N = 2,2'-bipyridine (bpy, in Ru-ART-1 and Ru-ART-4), 1,10-phenanthroline (phen, in Ru-ART-2 and Ru-ART-5) and 4,7-diphenyl-1,10-phenanthroline (DIP, in Ru-ART-3 and Ru-ART-6)), were synthesized and characterized. Among them, Ru-ART-1-3 and Ru-ART-4-6 carry one and two ART moieties, respectively. Ru-ART-3 and Ru-ART-6 exhibit better cytotoxicity among six Ru(II)-ART conjugates. These two complexes can be effectively taken up by human cervical carcinoma (HeLa) cells. In addition, they selectively kill cancer cell lines while mildly affect normal cells. Mechanism studies have shown that HeLa cells treated with Ru-ART-3 and Ru-ART-6 show typical apoptotic characteristics (morphology changes, mitochondrial dysfunction, caspase cascade, etc.). On the other hand, the up regulation of Beclin-1 and conversion of LC3-I to LC3-II note the appearance of autophagy. As a result, Ru-ART-3 and Ru-ART-6 induce autophagy-dependent cell apoptosis via mitochondrial dysfunction and reactive oxygen species (ROS) accumulation. In this work, six artesunate (ART) conjugated ruthenium(II) complexes (Ru(II)-ART conjugates) have been synthesized and characterized. Among them, Ru-ART-3 and Ru-ART-6 exhibit better cytotoxicity. Mechanism studies have shown that HeLa cells treated with Ru-ART-3 and Ru-ART-6 show typical apoptotic characteristics (morphology changes, mitochondrial dysfunction, caspase cascade, etc.). On the other hand, the up regulation of Beclin-1 and conversion of LC3-I to LC3-II note the appearance of autophagy.
Collapse
Affiliation(s)
- Bi-Chun Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China
| | - Jun-Jian Lu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China
| | - Ning Jiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China
| | - Xiu-Rong Ma
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China
| | - Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China.
| |
Collapse
|
19
|
da Silva MM, Ribeiro GH, de Camargo MS, Ferreira AG, Ribeiro L, Barbosa MIF, Deflon VM, Castelli S, Desideri A, Corrêa RS, Ribeiro AB, Nicolella HD, Ozelin SD, Tavares DC, Batista AA. Ruthenium(II) Diphosphine Complexes with Mercapto Ligands That Inhibit Topoisomerase IB and Suppress Tumor Growth In Vivo. Inorg Chem 2021; 60:14174-14189. [PMID: 34477373 DOI: 10.1021/acs.inorgchem.1c01539] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ruthenium(II) complexes (Ru1-Ru5), with the general formula [Ru(N-S)(dppe)2]PF6, bearing two 1,2-bis(diphenylphosphino)ethane (dppe) ligands and a series of mercapto ligands (N-S), have been developed. The combination of these ligands in the complexes endowed hydrophobic species with high cytotoxic activity against five cancer cell lines. For the A549 (lung) and MDA-MB-231 (breast) cancer cell lines, the IC50 values of the complexes were 288- to 14-fold lower when compared to cisplatin. Furthermore, the complexes were selective for the A549 and MDA-MB-231 cancer cell lines compared to the MRC-5 nontumor cell line. The multitarget character of the complexes was investigated by using calf thymus DNA (CT DNA), human serum albumin, and human topoisomerase IB (hTopIB). The complexes potently inhibited hTopIB. In particular, complex [Ru(dmp)(dppe)2]PF6 (Ru3), bearing the 4,6-diamino-2-mercaptopyrimidine (dmp) ligand, effectively inhibited hTopIB by acting on both the cleavage and religation steps of the catalytic cycle of this enzyme. Molecular docking showed that the Ru1-Ru5 complexes have binding affinity by active sites on the hTopI and hTopI-DNA, mainly via π-alkyl and alkyl hydrophobic interactions, as well as through hydrogen bonds. Complex Ru3 displayed significant antitumor activity against murine melanoma in mouse xenograph models, but this complex did not damage DNA, as revealed by Ames and micronucleus tests.
Collapse
Affiliation(s)
- Monize M da Silva
- Departamento de Química, Universidade Federal de São Carlos, CP 676, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Gabriel H Ribeiro
- Departamento de Química, Universidade Federal de São Carlos, CP 676, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Mariana S de Camargo
- Departamento de Química, Universidade Federal de São Carlos, CP 676, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Antônio G Ferreira
- Departamento de Química, Universidade Federal de São Carlos, CP 676, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Leandro Ribeiro
- Departamento de Química, Universidade Federal de São Carlos, CP 676, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Marília I F Barbosa
- Departamento de Química, Universidade Federal de São Carlos, CP 676, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Victor M Deflon
- Instituto de Química de São Carlos, Universidade de São Paulo, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Silvia Castelli
- Dipartimento di Biologia, Università Tor Vergata di Roma, 00133 Rome, Italy
| | | | - Rodrigo S Corrêa
- Departamento de Química, Universidade Federal de Ouro Preto, CEP 35400-000 Ouro Preto, Minas Gerais, Brazil
| | - Arthur B Ribeiro
- Universidade de Franca, CEP 14404-600, Franca, São Paulo, Brazil
| | | | - Saulo D Ozelin
- Universidade de Franca, CEP 14404-600, Franca, São Paulo, Brazil
| | - Denise C Tavares
- Universidade de Franca, CEP 14404-600, Franca, São Paulo, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, CP 676, CEP 13565-905 São Carlos, São Paulo, Brazil
| |
Collapse
|
20
|
Cyclometalated Ru(II) β-carboline complexes induce cell cycle arrest and apoptosis in human HeLa cervical cancer cells via suppressing ERK and Akt signaling. J Biol Inorg Chem 2021; 26:793-808. [PMID: 34459988 DOI: 10.1007/s00775-021-01894-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
Two new cyclometalated Ru(II)-β-carboline complexes, [Ru(dmb)2(Cl-Ph-βC)](PF6) (dmb = 4,4'-dimethyl-2,2'-bipyridine; Cl-Ph-βC = Cl-phenyl-9H-pyrido[3,4-b]indole; RuβC-3) and [Ru(bpy)2(Cl-Ph-βC)](PF6) (bpy = 2,2'-bipyridine; RuβC-4) were synthesized and characterized. The Ru(II) complexes display high cytotoxicity against HeLa cells, the stabilized human cervical cancer cell, with IC50 values of 3.2 ± 0.4 μM (RuβC-3) and 4.1 ± 0.6 μM (RuβC-4), which were considerably lower than that of non-cyclometalated Ru(II)-β-carboline complex [Ru(bpy)2(1-Py-βC)] (PF6)2 (61.2 ± 3.9 μM) by 19- and 15-folds, respectively. The mechanism studies indicated that both Ru(II) complexes could significantly inhibit HeLa cell migration and invasion, and effectively induce G0/G1 cell cycle arrest. The new Ru(II) complexes could also trigger apoptosis through activating caspase-3 and poly (ADP-ribose) polymerase (PARP), increasing the Bax/Bcl-2 ratio, enhancing reactive oxygen species (ROS) generation, decreasing mitochondrial membrane potential (MMP), and inducing cytochrome c release from mitochondria. Further research revealed that RuβC-3 could deactivate the ERK/Akt signaling pathway thus inhibiting HeLa cell invasion and migration, and inducing apoptosis. In addition, RuβC-3-induced apoptosis in HeLa cells was closely associated with the increase of intracellular ROS levels, which may act as upstream factors to regulate ERK and Akt pathways. More importantly, RuβC-3 exhibited low toxicity on both normal BEAS-2B cells in vitro and zebrafish embryos in vivo. Consequently, the developed Ru(II) complexes have great potential on developing novel low-toxic anticancer drugs.
Collapse
|
21
|
Zhu X, Li Z, Ji X, Chen Q, Wu S, Gao E, Zhu M. Two new lanthanide complexes with 5-(Pyrazol-1-yl)nicotinic acid: Structures and their anti-cancer properties. J Inorg Biochem 2021; 222:111505. [PMID: 34144467 DOI: 10.1016/j.jinorgbio.2021.111505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/29/2021] [Accepted: 05/30/2021] [Indexed: 02/08/2023]
Abstract
Two new lanthanide complexes [PrL2(EA)2]NO3 (complex 1) and [SmL2(EA)2]NO3 (complex 2) (H2L = 5-(Pyrazol-1-yl)nicotinic acid, EA = CH3CH2OH) were synthesized. The structures were characterized by single crystal X-ray and elemental analysis. The interaction between the complex and fish sperm DNA(FS-DNA) was monitored using ultraviolet and fluorescence spectroscopy, and the binding constants were determined. Both complexes showed the ability to effectively bind DNA, and the molecular docking technology was used to simulate the binding of the complex and DNA. In addition, through the annexin V-Fluorescein Isothiocyanate(FITC)/ Propidium Iodide (PI) test experiment, tetrazollium [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(MTT) in vitro test, and cell morphology apoptosis studies, it was shown that the complex can effectively induce HeLa tumor cell apoptosis. Compared with cisplatin and complex, complex 1 shows significant cancer cell inhibition, and we hope that this new type of complex will open up new ways for the next generation of drugs in biomedical applications.
Collapse
Affiliation(s)
- Xiaopeng Zhu
- International Key Laboratory of Liaoning Inorganic Molecule-Based Chemical and Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Zhipeng Li
- International Key Laboratory of Liaoning Inorganic Molecule-Based Chemical and Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Xiaoxi Ji
- International Key Laboratory of Liaoning Inorganic Molecule-Based Chemical and Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Qing Chen
- International Key Laboratory of Liaoning Inorganic Molecule-Based Chemical and Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Shuangyan Wu
- International Key Laboratory of Liaoning Inorganic Molecule-Based Chemical and Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Enjun Gao
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan 114051, PR China
| | - Mingchang Zhu
- International Key Laboratory of Liaoning Inorganic Molecule-Based Chemical and Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China; Key Laboratory of Resource Chemical Technology and Materials, (Ministry of Education), Shenyang University Chemical Technology, Shenyang 110142, PR China.
| |
Collapse
|
22
|
Velozo-Sa VS, Oliveira RM, Leite CM, Cominetti MR, Barbosa IM, Silva FL, Martins Feitosa N, Schultz MS, Batista AA. Scavenging capacity and cytotoxicity of new Ru(II)-diphosphine/α-amino acid complexes. Polyhedron 2021. [DOI: 10.1016/j.poly.2021.115169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
Nayeem N, Contel M. Exploring the Potential of Metallodrugs as Chemotherapeutics for Triple Negative Breast Cancer. Chemistry 2021; 27:8891-8917. [PMID: 33857345 DOI: 10.1002/chem.202100438] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Indexed: 12/11/2022]
Abstract
This review focuses on studies of coordination and organometallic compounds as potential chemotherapeutics against triple negative breast cancer (TNBC) which has one of the poorest prognoses and worst survival rates from all breast cancer types. At present, chemotherapy is still the standard of care for TNBC since only one type of targeted therapy has been recently developed. References for metal-based compounds studied in TNBC cell lines will be listed, and those of metal-specific reviews, but a detailed overview will also be provided on compounds studied in vivo (mostly in mice models) and those compounds for which some preliminary mechanistic data was obtained (in TNBC cell lines and tumors) and/or for which bioactive ligands have been used. The main goal of this review is to highlight the most promising metal-based compounds with potential as chemotherapeutic agents in TNBC.
Collapse
Affiliation(s)
- Nazia Nayeem
- Brooklyn College Cancer Center BCCC-CURE, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Biology PhD Program, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA
| | - Maria Contel
- Brooklyn College Cancer Center BCCC-CURE, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Biology PhD Program, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA.,Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA.,University of Hawaii Cancer Center, 701 Ilalo St, Honolulu, Hawaii, 96813, USA
| |
Collapse
|
24
|
Travassos IO, Mello-Andrade F, Caldeira RP, Pires WC, da Silva PFF, Correa RS, Teixeira T, Martins-Oliveira A, Batista AA, de Silveira-Lacerda EP. Ruthenium (II)/allopurinol complex inhibits breast cancer progression via multiple targets. J Biol Inorg Chem 2021; 26:385-401. [PMID: 33837856 DOI: 10.1007/s00775-021-01862-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/08/2021] [Indexed: 12/27/2022]
Abstract
Metal complexes based on ruthenium have established excellent activity with less toxicity and great selectivity for tumor cells. This study aims to assess the anticancer potential of ruthenium(II)/allopurinol complexes called [RuCl2(allo)2(PPh3)2] (1) and [RuCl2(allo)2(dppb)] (2), where allo means allopurinol, PPh3 is triphenylphosphine and dppb, 1,4-bis(diphenylphosphino)butane. The complexes were synthesized and characterized by elemental analysis, IR, UV-Vis and NMR spectroscopies, cyclic voltammetry, molar conductance measurements, as well as the X-ray crystallographic analysis of complex 2. The antitumor effects of compounds were determined by cytotoxic activity and cellular and molecular responses to cell death mechanisms. Complex 2 showed good antitumor profile prospects because in addition to its cytotoxicity, it causes cell cycle arrest, induction of DNA damage, morphological and biochemical alterations in the cells. Moreover, complex 2 induces cell death by p53-mediated apoptosis, caspase activation, increased Beclin-1 levels and decreased ROS levels. Therefore, complex 2 can be considered a suitable compound in antitumor treatment due to its cytotoxic mechanism.
Collapse
Affiliation(s)
- Ingrid O Travassos
- Laboratório de Genética Molecular E Citogenética Humana, sala 213, Departamento de Genética, Instituto de Ciências Biológicas I, Campus Samambaia, Universidade Federal de Goiás, Avenida Esperança, s/n, Cx Postal: 131, Goiânia, Goiás, CEP 74690-900, Brazil
| | - Francyelli Mello-Andrade
- Laboratório de Genética Molecular E Citogenética Humana, sala 213, Departamento de Genética, Instituto de Ciências Biológicas I, Campus Samambaia, Universidade Federal de Goiás, Avenida Esperança, s/n, Cx Postal: 131, Goiânia, Goiás, CEP 74690-900, Brazil.,Department of Chemistry, Federal Institute of Education, Science and Technology of Goiás, Goiânia, Goiás, 74055-110, Brazil
| | - Raíssa P Caldeira
- Laboratório de Genética Molecular E Citogenética Humana, sala 213, Departamento de Genética, Instituto de Ciências Biológicas I, Campus Samambaia, Universidade Federal de Goiás, Avenida Esperança, s/n, Cx Postal: 131, Goiânia, Goiás, CEP 74690-900, Brazil
| | - Wanessa C Pires
- Laboratório de Genética Molecular E Citogenética Humana, sala 213, Departamento de Genética, Instituto de Ciências Biológicas I, Campus Samambaia, Universidade Federal de Goiás, Avenida Esperança, s/n, Cx Postal: 131, Goiânia, Goiás, CEP 74690-900, Brazil
| | - Paula F F da Silva
- Laboratório de Genética Molecular E Citogenética Humana, sala 213, Departamento de Genética, Instituto de Ciências Biológicas I, Campus Samambaia, Universidade Federal de Goiás, Avenida Esperança, s/n, Cx Postal: 131, Goiânia, Goiás, CEP 74690-900, Brazil
| | - Rodrigo S Correa
- Department of Chemistry, Federal University of Ouro Preto-UFOP, Ouro Preto, MG, 35400-000, Brazil
| | - Tamara Teixeira
- Department of Chemistry, Federal University of Ouro Preto-UFOP, Ouro Preto, MG, 35400-000, Brazil
| | | | - Alzir A Batista
- Department of Chemistry, Federal University of Sao Carlos-UFSCar, Sao Carlos, SP, 13565-905, Brazil
| | - Elisângela P de Silveira-Lacerda
- Laboratório de Genética Molecular E Citogenética Humana, sala 213, Departamento de Genética, Instituto de Ciências Biológicas I, Campus Samambaia, Universidade Federal de Goiás, Avenida Esperança, s/n, Cx Postal: 131, Goiânia, Goiás, CEP 74690-900, Brazil.
| |
Collapse
|
25
|
Kamal NAMA, Abdulmalek E, Fakurazi S, Cordova KE, Abdul Rahman MB. Surface peptide functionalization of zeolitic imidazolate framework-8 for autonomous homing and enhanced delivery of chemotherapeutic agent to lung tumor cells. Dalton Trans 2021; 50:2375-2386. [PMID: 33555001 DOI: 10.1039/d1dt00116g] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chemotherapeutic agents used in treating certain cancer types operate in a non-selective manner tending to accumulate in normal, healthy tissue when high doses are used. To mitigate the toxicity effect resulting from this, there is an urgent need to develop active nano delivery systems capable of regulating optimal doses specifically to cancer cells without harming adjacent normal cells. Herein, we report a versatile nanoparticle - zeolitic imidazolate framework-8 (nZIF-8) - that is loaded with a chemotherapeutic agent (gemcitabine; GEM) and surface-functionalized with an autonomous homing system (Arg-Gly-Asp peptide ligand; RGD) via a straightforward, one-pot solvothermal reaction. Successful functionalization of the surface of nZIF-8 loaded GEM (GEM⊂nZIF-8) with RGD was proven by spectroscopic and electron microscopy techniques. This surface-functionalized nanoparticle (GEM⊂RGD@nZIF-8) exhibited enhanced uptake in human lung cancer cells (A549), compared with non-functionalized GEM⊂nZIF-8. The GEM⊂RGD@nZIF-8, experienced not only efficient uptake within A549, but also induced obvious cytotoxicity (75% at a concentration of 10 μg mL-1) and apoptosis (62%) after 48 h treatment when compared to the nanoparticle absent of the RGD homing system (GEM⊂nZIF-8). Most importantly, this surface-functionalized nanoparticle was more selective towards lung cancer cells (A549) than normal human lung fibroblast cells (MRC-5) with a selectivity index (SI) of 3.98. This work demonstrates a new one-pot strategy for realizing a surface-functionalized zeolitic imidazolate framework that actively targets cancer cells via an autonomous homing peptide system to deliver a chemotherapeutic payload effectively.
Collapse
Affiliation(s)
- Nurul Akmarina Mohd Abdul Kamal
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia. and UPM-MAKNA Cancer Laboratory, Institute of Bioscience, UPM, Serdang 43400, Selangor, Malaysia and Faculty of Chemical and Process Engineering Technology, Universiti Malaysia Pahang (UMP), Pekan 26600, Pahang, Malaysia
| | - Emilia Abdulmalek
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia.
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, UPM, Serdang 43400, Selangor, Malaysia
| | - Kyle E Cordova
- Materials Discovery Research Unit, Advanced Research Centre, Royal Scientific Society, Amman 11941, Jordan.
| | - Mohd Basyaruddin Abdul Rahman
- Integrated Chemical BioPhysics Research, Department of Chemistry, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia. and UPM-MAKNA Cancer Laboratory, Institute of Bioscience, UPM, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
26
|
Teixeira TM, Arraes IG, Abreu DC, Oliveira KM, Correa RS, Batista AA, Braunbeck T, de Paula Silveira Lacerda E. Ruthenium complexes show promise when submitted to toxicological safety tests using alternative methodologies. Eur J Med Chem 2021; 216:113262. [PMID: 33711764 DOI: 10.1016/j.ejmech.2021.113262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/30/2021] [Accepted: 01/31/2021] [Indexed: 12/30/2022]
Abstract
The number of cancer cases continues to increase worldwide, and unfortunately the main systemic treatments available have numerous of side effects. Ruthenium complexes have shown to be promising chemotherapeutic agents, since they present low toxicity and are more selective for tumor tissues. We report the synthesis, characterization and biological properties of two new ruthenium (II) complexes containing Lapachol and Lawsone as ligands: (1) [Ru(Law)(dppb)(phen)]PF6 and (2) [Ru(Lap)(dppb)(phen)]PF6, where Law = Lawsone, Lap = Lapachol, dppb = 1,4-bis(diphenylphosphine)butane and phen = 1,10-phenanthroline. The ability of the complexes (1) and (2) to interact with CT-DNA (Calf Thymus) was investigated, and the results indicate that the complexes have shown a weak interaction with this macromolecule. Complexes (1) and (2) showed a moderate interaction with BSA, via a spontaneous process with the involvement of van der Waals and hydrogen bond interactions. Both complexes were tested against human lung cancer cell lines, chronic human myeloid leukemia, murine melanoma and human cervical and non-tumoral murine fibroblast adenocarcinoma, human lung fibroblasts and monkey kidney epithelia. The potential for cytotoxicity was tested out using the MTT assay and the neutral red test, to calculate inhibitory concentrations (IC50) and selectivity indices (IS). Both complexes showed a higher selectivity index of 1.17 and 10.91, respectively, for the HeLa tumor line. Studies of toxicological evaluation, using the micronucleus test and the comet assay against non-tumor cells, as well as an assessment of the potential for acute toxicity and neurotoxicity in zebrafish (Danio rerio). In the in vitro micronucleus test, complex (1) showed the least genotoxic potential, and in the in vitro comet assay both compounds had revealed a genotoxic potential at 0.5 and 1.0 mg L-1, with no difference between 24 h and 48 h exposure times. In the acute toxicity tests on zebrafish embryos, complex (1) showed sublethal effects such as decreased blood circulation and heartbeat rate, which were less pronounced than with complex (2). In contrast to complex 2, which caused lethality even before 48h, complex (1) did not cause the death of the embryos at concentrations up to (2.0 mg L-1). Complex (2) also lead to a delay in the embryo. Cell based in vitro methods thus proved able to provide specific toxicological data, allowing a significant reduction in ∖animal experimentation. Given that in vitro tests cannot completely replace animal tests, the use of less advanced developmental stages such as zebrafish embryos, which - at least in the European Union - are not regarded protected, could be shown to be an excellent alternative for testing with, e.g., mammals.
Collapse
Affiliation(s)
- Thallita Monteiro Teixeira
- Instituto de Ciências Biológicas, Universidade Federal de Goiás (UFG), CEP 74045-155, Goiânia, GO, Brazil; Aquatic Ecology and Toxicology, Center for Organismal Studies, University of Heidelberg, D-69117, Heidelberg, Germany.
| | - Isabela Gasparini Arraes
- Instituto de Ciências Biológicas, Universidade Federal de Goiás (UFG), CEP 74045-155, Goiânia, GO, Brazil
| | - Davi Carvalho Abreu
- Instituto de Ciências Biológicas, Universidade Federal de Goiás (UFG), CEP 74045-155, Goiânia, GO, Brazil
| | - Katia M Oliveira
- Departamento de Química, Universidade Federal de São Carlos (UFSCar), CP 676, CEP 13565-905, São Carlos, SP, Brazil; Departamento de Química, ICEB, Universidade Federal de Ouro Preto (UFOP), CEP 35400-000, Ouro Preto, MG, Brazil
| | - Rodrigo S Correa
- Departamento de Química, ICEB, Universidade Federal de Ouro Preto (UFOP), CEP 35400-000, Ouro Preto, MG, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos (UFSCar), CP 676, CEP 13565-905, São Carlos, SP, Brazil
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology, Center for Organismal Studies, University of Heidelberg, D-69117, Heidelberg, Germany
| | | |
Collapse
|
27
|
Lapachol in the Design of a New Ruthenium(II)-Diphosphine Complex as a Promising Anticancer Metallodrug. J Inorg Biochem 2020; 214:111289. [PMID: 33137682 DOI: 10.1016/j.jinorgbio.2020.111289] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/08/2020] [Accepted: 10/17/2020] [Indexed: 12/26/2022]
Abstract
The preparation of two new Ru(II)/diphosphine complexes containing Lapachol (Lap) and Lawsone (Law): (1) [Ru(Lap)(dppm)2]PF6 and (2) [Ru(Law)(dppm)2]PF6, where dppm = bis(diphenylphosphino)methane, is reported here. The complexes were synthetized and fully characterized by elemental analyses, molar conductivity, UV-Vis, IR, 31P{1H}, 1H and 13C NMR, and the crystal structure of the complex (1) was determined by X-ray diffraction. Complexes (1) and (2) showed high in vitro cytotoxicity against four cancer cells (MDA-MB-231, MCF-7, A549 and DU-145), with IC50 values in the micromolar range (0.03 to 2.70 μM). Importantly, complexes (1) and (2) were more active than the cisplatin, the drug used as a reference in the cytotoxic assays. Moreover, complex (1) showed high selectivity to triple-negative breast cancer cells (MDA-MB-231). Studies of the mechanism of action in MDA-MB-231 cancer cells showed that complex (1) inhibits cell migration, colony formation, and induces cell cycle arrest and apoptosis by activation of the mitochondrial pathway through the loss of mitochondrial membrane potential (ΔΨm). Furthermore, complex (1) induces ROS (Reactive Oxygen Species) generation in MDA-MB-231 cells, which can cause DNA damage. Finally, complexes (1) and (2) interact with DNA by minor grooves and show a moderate interaction with BSA (Bovine Serum Albumin), with the involvement of hydrophobic interactions. Essentially, Ru(II)/diphosphine-naphthoquinone complexes have remarkable cytotoxic effects with high selectivity to triple-negative breast cancer (MDA-MB-231) and could be promising anticancer candidates for cancer treatment. SYNOPSIS: The naphthoquinones Lapachol and Lawsone can form new ruthenium compounds with promising anticancer properties.
Collapse
|
28
|
Trigueiro NSDS, Canedo A, Braga DLDS, Luchiari AC, Rocha TL. Zebrafish as an Emerging Model System in the Global South: Two Decades of Research in Brazil. Zebrafish 2020; 17:412-425. [PMID: 33090089 DOI: 10.1089/zeb.2020.1930] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The zebrafish (Danio rerio) is an emerging model system in several research areas worldwide, especially in the Global South. In this context, the present study revised the historical use and trends of zebrafish as experimental models in Brazil. The data concerning the bibliometric parameters, research areas, geographic distribution, experimental design, zebrafish strain, and reporter lines, as well as recent advances were revised. In addition, the comparative trends of Brazilian and global research were discussed. Revised data showed the rapid growth of Brazilian scientific production using zebrafish as a model, especially in three main research areas (Neuroscience &and Behavior, Pharmacology and Toxicology, and Environment/Ecology). Studies were conducted in 19 Brazilian states (70.37%), confirming the wide geographic distribution and importance of zebrafish research. Results indicated that research related to toxicological approaches are widespread in Global South countries such as Brazil. Studies were performed mainly using in vivo tests (89.58%) with adult fish (59.75%) and embryos (30.67%). Moreover, significant research gaps and recommendations for future research are presented. The present study shows that the zebrafish is a suitable vertebrate model system in the Global South.
Collapse
Affiliation(s)
- Nicholas Silvestre de Souza Trigueiro
- Laboratory of Environmental Biotechnology and Ecotoxicology, Department of Biosciences and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | - Aryelle Canedo
- Laboratory of Environmental Biotechnology and Ecotoxicology, Department of Biosciences and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | - Daniel Lôbo de Siqueira Braga
- Laboratory of Environmental Biotechnology and Ecotoxicology, Department of Biosciences and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | - Ana Carolina Luchiari
- Department of Physiology and Behavior, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Thiago Lopes Rocha
- Laboratory of Environmental Biotechnology and Ecotoxicology, Department of Biosciences and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
29
|
Ribeiro GH, Guedes APM, de Oliveira TD, de Correia CRSTB, Colina-Vegas L, Lima MA, Nóbrega JA, Cominetti MR, Rocha FV, Ferreira AG, Castellano EE, Teixeira FR, Batista AA. Ruthenium(II) Phosphine/Mercapto Complexes: Their in Vitro Cytotoxicity Evaluation and Actions as Inhibitors of Topoisomerase and Proteasome Acting as Possible Triggers of Cell Death Induction. Inorg Chem 2020; 59:15004-15018. [PMID: 32997499 DOI: 10.1021/acs.inorgchem.0c01835] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this paper, a series of new ruthenium complexes of the general formula [Ru(NS)(dpphpy)(dppb)]PF6 (Ru1-Ru3), where dpphpy = diphenyl-2-pyridylphosphine, NS ligands = 2-thiazoline-2-thiol (tzdt, Ru1), 2-mercaptopyrimidine (pySm, Ru2), and 4,6-diamino-2-mercaptopyrimidine (damp, Ru3), and dppb = 1,4-bis(diphenylphosphino)butane, were synthesized and characterized by elemental analysis, spectroscopic techniques (IR, UV/visible, and 1D and 2D NMR), and X-ray diffraction. In the characterization, the correlation between the phosphorus atoms and their respective aromatic hydrogen atoms of the compounds in the assignment stands outs, by 1H-31P HMBC experiments. The compounds show anticancer activities against A549 (lung) and MDA-MB-231 (breast) cancer cell lines, higher than the clinical drug cisplatin. All of the complexes are more cytotoxic against the cancer cell lines than against the MRC-5 (lung) and MCF-10A (breast) nontumorigenic human cell lines. For A549 tumor cells, cell cycle analysis upon treatment with Ru2 showed that it inhibits the mitotic phase because arrest was observed in the Sub-G1 phase. Additionally, the compound induces cell death by an apoptotic pathway in a dose-dependent manner, according to annexin V-PE assay. The multitargeted character of the compounds was investigated, and the biomolecules were DNA, topoisomerase IB, and proteasome, as well as the fundamental biomolecule in the pharmacokinetics of drugs, human serum albumin. The experimental results indicate that the complexes do not target DNA in the cells. At low concentrations, the compounds showed the ability to partially inhibit the catalytic activity of topoisomerase IB in the process of relaxation of the DNA plasmid. Among the complexes assayed in cultured cells, complex Ru3 was able to diminish the proteasomal chymotrypsin-like activity to a greater extent.
Collapse
Affiliation(s)
- Gabriel H Ribeiro
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Adriana P M Guedes
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Tamires D de Oliveira
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Camila R S T B de Correia
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Legna Colina-Vegas
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil.,Instituto de Química, Universidade Federal do Rio Grande do Sul, CP 15003, 91501-970 Porto Alegre, Rio Grande do Sul, Brazil
| | - Mauro A Lima
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Joaquim A Nóbrega
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Márcia R Cominetti
- Departamento de Gerontologia, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo Brazil
| | - Fillipe V Rocha
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Antônio G Ferreira
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Eduardo E Castellano
- Instituto de Física de São Carlos, Universidade de São Paulo, CEP 13560-970 São Carlos, São Paulo, Brazil
| | - Felipe R Teixeira
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| |
Collapse
|
30
|
de Lima AP, Almeida MAP, Mello-Andrade F, de Castro Pereira F, Pires WC, Abreu DC, de Souza Velozo-Sá V, Batista AA, de Paula Silveira-Lacerda E. Ru(II)-Based Amino Acid Complexes Show Promise for Leukemia Treatment: Cytotoxicity and Some Light on their Mechanism of Action. Biol Trace Elem Res 2020; 197:123-131. [PMID: 31773484 DOI: 10.1007/s12011-019-01976-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/01/2019] [Indexed: 12/20/2022]
Abstract
Ruthenium is attracting considerable interest as the basis for new compounds to treat diseases, and studies have shown that complexes with different structures have significant antineoplastic and antimetastatic potential against several types of tumors, including tumors resistant to cisplatin drugs. We examined the cytotoxic, genotoxic, and pro-apoptotic activities of six ruthenium complexes containing amino acid with general formulation [Ru(AA)(bipy)(dppb)]PF6, where AA = amino acid (alanine, glycine, leucine, lysine, methionine, or tryptophan); bipy = 2,2´-bipyridine; and dppb = [1,4-bis(diphenylphosphine)butane], against A549 (lung carcinoma) and K562 (chronic myelogenous leukemia) cancer cells. The results show that the ruthenium complexes tested were able to induce cytotoxicity in A549 and K562 cancer cells. Complex 1 containing alanine inhibited the cell viability of A549 and K562 tumor cells by inducing apoptosis, as evidenced by an increased number of Annexin V-positive cells and the induction of DNA damage and cell cycle arrest. Complex 1 was able to induce caspase-mediated apoptosis in K562 cells through the mitochondrial dysfunction, the upregulation of apoptotic genes, and the downregulation of Bcl2 anti-apoptotic gene. Besides being cytotoxic to K562 and A549 cells, ruthenium complex containing alanine shows low cytotoxicity and genotoxicity against non-tumor cells. These results suggest that the ruthenium (II) complex is a potential safe and efficient antineoplastic candidate for leukemia treatment.
Collapse
Affiliation(s)
- Aliny Pereira de Lima
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
- Faculty of Brazil Institute (FIBRA), Anápolis, Goiás, 75133-050, Brazil
| | | | - Francyelli Mello-Andrade
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
- Department of Chemistry, Federal Institute of Education, Science and Technology of Goiás, Goiânia, Goiás, 74055-110, Brazil
| | - Flávia de Castro Pereira
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Wanessa Carvalho Pires
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Davi Carvalho Abreu
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Vivianne de Souza Velozo-Sá
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Alzir Azevedo Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13565-905, Brazil
| | - Elisângela de Paula Silveira-Lacerda
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil.
| |
Collapse
|
31
|
Toupin NP, Nadella S, Steinke SJ, Turro C, Kodanko JJ. Dual-Action Ru(II) Complexes with Bulky π-Expansive Ligands: Phototoxicity without DNA Intercalation. Inorg Chem 2020; 59:3919-3933. [PMID: 32096986 DOI: 10.1021/acs.inorgchem.9b03585] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report the synthesis and photochemical and biological characterization of Ru(II) complexes containing π-expansive ligands derived from dimethylbenzo[i]dipyrido[3,2-a:2',3'-c]phenazine (Me2dppn) adorned with flanking aryl substituents. Late-stage Suzuki couplings produced Me2dppn ligands substituted at the 10 and 15 positions with phenyl (5), 2,4-dimethylphenyl (6), and 2,4-dimethoxyphenyl (7) groups. Complexes of the general formula [Ru(tpy)(L)(py)](PF6)2 (8-10), where L = 4-7, were characterized and shown to have dual photochemotherapeutic (PCT) and photodynamic therapy (PDT) behavior. Quantum yields for photodissociation of monodentate pyridines from 8-10 were about 3 times higher than that of parent complex [Ru(tpy)(Me2dppn)(py)](PF6)2 (1), whereas quantum yields for singlet oxygen (1O2) production were ∼10% lower than that of 1. Transient absorption spectroscopy indicates that 8-10 possess long excited state lifetimes (τ = 46-50 μs), consistent with efficient 1O2 production through population and subsequent decay of ligand-centered 3ππ* excited states. Complexes 8-10 displayed greater lipophilicity relative to 1 and association to DNA but do not intercalate between the duplex base pairs. Complexes 1 and 8-10 showed photoactivated toxicity in breast and prostate cancer cell lines with phototherapeutic indexes, PIs, as high as >56, where the majority of cell death was achieved 4 h after treatment with Ru(II) complexes and light. Flow cytometric data and rescue experiments were consistent with necrotic cell death mediated by the production of reactive oxygen species, especially 1O2. Collectively, this study confirms that DNA intercalation by Ru(II) complexes with π-expansive ligands is not required to achieve photoactivated cell death.
Collapse
Affiliation(s)
- Nicholas P Toupin
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Sandeep Nadella
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Sean J Steinke
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Claudia Turro
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jeremy J Kodanko
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States.,Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| |
Collapse
|
32
|
Correa RS, Bomfim LM, Oliveira KM, Moreira DR, Soares MB, Ellena J, Bezerra DP, Batista AA. Ru(II) complexes containing uracil nucleobase analogs with cytotoxicity against tumor cells. J Inorg Biochem 2019; 198:110751. [DOI: 10.1016/j.jinorgbio.2019.110751] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/22/2019] [Accepted: 06/05/2019] [Indexed: 01/31/2023]
|