1
|
de Alencar FMS, Gouveia FS, Oliveira GDFSD, Andrade AL, Vasconcelos MAD, Ayala AP, Gondim ACS, Carvalho IMMD, Moraes CAF, Palmeira-Mello MV, Batista AA, Lopes LGDF, Sousa EHS. Terpyridine-based ruthenium complexes containing a 4,5-diazafluoren-9-one ligand with light-driven enhancement of biological activity. Dalton Trans 2024. [PMID: 39686803 DOI: 10.1039/d4dt02562h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
There has been growing effort in the scientific community to develop new antibiotics to address the major threat of bacterial resistance. One promising approach is the use of metal complexes that provide broader opportunities. Among these systems, polypyridine-ruthenium(II) complexes have received particular attention as drug candidates. Here, we prepared two new ruthenium(II) complexes with the formulation [Ru(DFO)(phtpy-R)Cl](PF6), where phtpy = 4'-phenyl-2,2':6',2''-terpyridine; R = -H(MPD1), -CH3(MPD2); and DFO = 4,5-diazafluoren-9-one, and investigated their chemical, biochemical and antibacterial activities. These compounds exhibit photoreactivity and produce reactive oxygen species (ROSs). Photogeneration of singlet oxygen (1O2) was measured in acetonitrile with significant quantum yields using blue light, Φ = 0.40 and 0.39 for MDP1 and MPD2, respectively. Further studies have shown that MPD1 and MPD2 can generate superoxide radicals. Antibacterial assays demonstrated a significant enhancement in MIC (minimum inhibitory concentration) upon blue light irradiation (>32-fold), with MICs of 15.6 μg mL-1 (S. aureus, ATCC 700698) and 3.9 μg mL-1 (S. epidermidis, ATCC 35984) for both metal complexes. Interestingly, an MIC of 15.6 μg mL-1 for MPD1 and MPD2 was observed against S. epidermidis ATCC 12228 under red light irradiation. The latter results are encouraging, considering that red light penetrates deeper into the skin. In addition, no significant cytotoxicity was observed in some mammalian cells, even upon light irradiation, supporting their potential safety. Altogether, these data show evidence of the potential use of these compounds as antimicrobial photodynamic therapeutic agents, enriching our arsenal to combat this worldwide bacterial threat.
Collapse
Affiliation(s)
| | - Florencio Sousa Gouveia
- Group of Bioinorganic, Department of Organic and Inorganic Chemistry, Federal University of Ceara, Fortaleza, Brazil.
| | | | - Alexandre Lopes Andrade
- Integrated Biomolecular Laboratory, Department of Pathology and Legal Medicine, Federal University of Ceara, Fortaleza, Brazil
| | | | | | - Ana Claudia Silva Gondim
- Group of Bioinorganic, Department of Organic and Inorganic Chemistry, Federal University of Ceara, Fortaleza, Brazil.
| | | | | | - Marcos V Palmeira-Mello
- Department of Chemistry, Federal University of São Carlos, 13565-905, São Carlos, SP, Brazil
| | - Alzir Azevedo Batista
- Department of Chemistry, Federal University of São Carlos, 13565-905, São Carlos, SP, Brazil
| | - Luiz Gonzaga de França Lopes
- Group of Bioinorganic, Department of Organic and Inorganic Chemistry, Federal University of Ceara, Fortaleza, Brazil.
| | - Eduardo Henrique Silva Sousa
- Group of Bioinorganic, Department of Organic and Inorganic Chemistry, Federal University of Ceara, Fortaleza, Brazil.
| |
Collapse
|
2
|
Gonzalo-Navarro C, Troyano AJ, Bermejo BGB, Organero JÁ, Massaguer A, Santos L, Rodríguez AM, Manzano BR, Durá G. Ru-terpyridine complexes containing clotrimazole as potent photoactivatable selective antifungal agents. J Inorg Biochem 2024; 260:112692. [PMID: 39151234 DOI: 10.1016/j.jinorgbio.2024.112692] [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: 06/17/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
The overuse of antimicrobial agents in medical and veterinary applications has led to the development of antimicrobial resistance in some microorganisms and this is now one of the major concerns in modern society. In this context, the use of transition metal complexes with photoactivatable properties, which can act as drug delivery systems triggered by light, could become a potent strategy to overcome the problem of resistance. In this work several Ru complexes with terpyridine ligands and the clotrimazole fragment, which is a potent antimycotic drug, were synthesized. The main goal was to explore the potential photoactivated activity of the complexes as antifungal agents and evaluate the effect of introducing different substituents on the terpyridine ligand. The complexes were capable of delivering the clotrimazole unit upon irradiation with visible light in a short period of time. The influence of the substituents on the photodissociation rate was explained by means of TD-DFT calculations. The complexes were tested against three different yeasts, which were selected based on their prevalence in fungal infections. The complex in which a carboxybenzene unit was attached to the terpyridine ligand showed the best activity against the three species under light, with minimal inhibitory concentration values of 0.88 μM and a phototoxicity index of 50 achieved. The activity of this complex was markedly higher than that of free clotrimazole, especially upon irradiation with visible light (141 times higher). The complexes were more active on yeast species than on cancer cell lines.
Collapse
Affiliation(s)
- Carlos Gonzalo-Navarro
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Ciencias y Tecnologías Químicas, Avda. C. J. Cela, 10, UCLM, Ciudad Real, Spain
| | - Antonio J Troyano
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Ciencias y Tecnologías Químicas, Avda. C. J. Cela, 10, UCLM, Ciudad Real, Spain
| | - Beatriz García-Béjar Bermejo
- Departamento de Química Analítica y Tecnología de los Alimentos, Ed. Marie Curie, Avenida C. J. Cela, s/n, UCLM, Ciudad Real, Spain
| | - Juan Ángel Organero
- Universidad de Castilla-La Mancha, Departamento de Química Física, Facultad de Ciencias Ambientales y Bioquímicas and INAMOL, 45071 Toledo, Spain
| | - Anna Massaguer
- Universitat de Girona, Departament de Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, 17003 Girona, Spain
| | - Lucía Santos
- Departamento de Q. Física, Facultad de Ciencias y Tecnologías Químicas, Avda. C. J. Cela, s/n, UCLM, Ciudad Real, Spain
| | - Ana M Rodríguez
- Departamento de Química Inorgánica, Orgánica y Bioquímica- IRICA, Escuela Técnica Superior de Ingenieros Industriales, Avda. C. J. Cela, 3, UCLM, Ciudad Real, Spain
| | - Blanca R Manzano
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Ciencias y Tecnologías Químicas, Avda. C. J. Cela, 10, UCLM, Ciudad Real, Spain
| | - Gema Durá
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Ciencias y Tecnologías Químicas, Avda. C. J. Cela, 10, UCLM, Ciudad Real, Spain.
| |
Collapse
|
3
|
Mandal AA, Upadhyay A, Mandal A, Nayak M, K MS, Mukherjee S, Banerjee S. Visible-Light-Responsive Novel Ru(II)-Metallo-Antibiotics with Potential Antibiofilm and Antibacterial Activity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28118-28133. [PMID: 38783713 DOI: 10.1021/acsami.4c02979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Growing challenges with antibiotic resistance pose immense challenges in combating microbial infections and biofilm prevention on medical devices. Lately, antibacterial photodynamic therapy (aPDT) is now emerging as an alternative therapy to overcome this problem. Herein, we synthesized and characterized four Ru(II)-complexes, viz., [Ru(ph-tpy)(bpy)Cl]PF6 (Ru1), [Ru(ph-tpy)(dpq)Cl]PF6 (Ru2), [Ru(ph-tpy)(dppz)Cl]PF6 (Ru3), and [Ru(ph-tpy)(dppn)Cl]PF6 (Ru4) (where 4'-phenyl-2,2':6',2″-terpyridine = ph-tpy; 2,2'-bipyridine = bpy; dipyrido[3,2-f:2',3'-h]quinoxaline = dpq; dipyrido[3,2-a:2',3'-c]phenazine = dppz; and Benzo[I]dipyrido[3,2-a:2',3'-c]phenazine = dppn), among which Ru2-Ru4 are novel. Octahedral geometry of the complexes with a RuN5Cl core was evident from the crystal structure of Ru2. Ru1-Ru4 showed an MLCT absorption band in the 450-600 nm region, useful for aPDT performances. Further, optimum triplet excited state energy and excellent photostability of Ru1-Ru4 made them good photosensitizers for aPDT. Ru1-Ru4 demonstrated enhanced antimicrobial activity on visible-light exposure (400-700 nm, 10 J cm-2), confirmed using different antibacterial assays. Mechanistic studies revealed that inhibition of bacterial growth was due to the generation of oxidative stress (via NADH oxidation and ROS generation) upon treatment with Ru2-Ru4, resulting in destruction of the bacterial wall. Ru2 performed best killing performance against both Gram-negative (Escherichia coli) and Gram-positive (Bacillus subtilis) bacteria when exposed to light. Ru2-Ru4, when coated on a polydimethylsiloxane (PDMS) disk, showed long-term reusability and durable antibiofilm properties. Molecular docking confirmed the efficient interaction of Ru2-Ru4 with FabH (regulates fatty acid biosynthesis of E. coli) and PgaB (gives structural stability and helps biofilm formation of E. coli), resulting in probable downregulation. In vivo studies with healthy Wistar rats confirmed the biocompatibility of Ru2. This study shows that these lead complexes (Ru2-Ru4) can be used as potent alternative antimicrobial agents in low concentrations toward bacterial eradication with photodynamic therapy (PDT).
Collapse
Affiliation(s)
- Arif Ali Mandal
- Department of Chemistry, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Anjali Upadhyay
- School of Biomedical Engineering, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Apurba Mandal
- Department of Chemistry, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Malay Nayak
- School of Biomedical Engineering, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Mohammad Sabeel K
- Department of Chemistry, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Samya Banerjee
- Department of Chemistry, IIT (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
4
|
Wrobel EC, Guimarães IDL, Wohnrath K, Oliveira ON. Effects induced by η 6-p-cymene ruthenium(II) complexes on Langmuir monolayers mimicking cancer and healthy cell membranes do not correlate with their toxicity. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184332. [PMID: 38740123 DOI: 10.1016/j.bbamem.2024.184332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
The mechanism of chemotherapeutic action of Ru-based drugs involves plasma membrane disruption and valuable insights into this process may be gained using cell membrane models. The interactions of a series of cytotoxic η6-p-cymene ruthenium(II) complexes, [Ru(η6-p-cymene)P(3,5-C(CH3)3-C6H3)3Cl2] (1), [Ru(η6-p-cymene)P(3,5-CH3-C6H3)3Cl2] (2), [Ru(η6-p-cymene)P(4-CH3O-3,5-CH3-C6H2)3Cl2] (3), and [Ru(η6-p-cymene)P(4-CH3O-C6H4)3Cl2] (4), were examined using Langmuir monolayers as simplified healthy and cancerous outer leaflet plasma membrane models. The cancerous membrane (CM1 and CM2) models contained either 40 % 1,2- dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) or 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 30 % cholesterol (Chol), 20 % 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE), and 10 % 1,2-dipalmitoyl-sn-glycero-3-phospho-l-serine (DPPS). Meanwhile, the healthy membrane (HM1 and HM2) models were composed of 60 % DPPC or DOPC, 30 % Chol and 10 % DPPE. The complexes affected surface pressure isotherms and decreased compressional moduli of cancerous and healthy membrane models, interacting with the monolayers headgroup and tails according to data from polarization-modulated infrared reflection absorption spectroscopy (PM-IRRAS). However, the effects did not correlate with the toxicity of the complexes to cancerous and healthy cells. Multidimensional projection technique showed that the complex (1) induced significant changes in the CM1 and HM1 monolayers, though it had the lowest cytotoxicity against cancer cells and is not toxic to healthy cells. Moreover, the most toxic complexes (2) and (4) were those that least affected CM2 and HM2 monolayers. The findings here support that the ruthenium complexes interact with lipids and cholesterol in cell membrane models, and their cytotoxic activities involve a multifaceted mode of action beyond membrane disruption.
Collapse
Affiliation(s)
- Ellen C Wrobel
- São Carlos Institute of Physics, University of São Paulo, CP 369, São Carlos, São Paulo, SP 13560-970, Brazil.
| | | | - Karen Wohnrath
- Department of Chemistry, Universidade Estadual de Ponta Grossa, Ponta Grossa, Paraná 84030-900, Brazil
| | - Osvaldo N Oliveira
- São Carlos Institute of Physics, University of São Paulo, CP 369, São Carlos, São Paulo, SP 13560-970, Brazil.
| |
Collapse
|
5
|
Jiang J, Chen Q, Huan T, Nie Y, Dai Z, Li D, Xu X, Lu J, Hu Z, Xu H. Comparative studies on in vitro antitumor activities and apoptosis-inducing effects of enantiomeric ruthenium(II) complexes. Dalton Trans 2023; 52:14338-14349. [PMID: 37431624 DOI: 10.1039/d3dt01584j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
On the basis of our previous comparative studies on the DNA binding of a pair of ruthenium(II) complex enantiomers, Δ-[Ru(bpy)2PBIP]2+ and Λ-[Ru(bpy)2PBIP]2+ {bpy = 2,2'-bipyridine, PBIP = 2-(4-bromophenyl)imidazo[4,5-f]1,10-phenanthroline}, in this study, their antitumor activities and mechanisms were further investigated comparatively. The cytotoxicity assay demonstrated that both the enantiomers exerted selective antiproliferative effects on cancer cell lines A2780 and PC3. Fluorescence localization experiments suggested that both the enantiomers effectively permeated the nucleus of HeLa cells and co-localized with DNA, resulting in their DNA damage and apoptosis. Flow cytometry experiments showed that the apoptosis was enhanced by increasing the concentration of each enantiomer. Western blotting analyses indicated that both extrinsic and intrinsic apoptosis pathways were activated by the two enantiomers. miRNA microarray analyses displayed that both the enantiomers up- and downregulated multiple miRNAs, some of which were predicted to be associated with carcinogenesis. The above experimental results also showed that the Δ-enantiomer exerted a more potent antitumor activity, a higher efficiency of entering cancer cells and a stronger apoptosis-inducing effect compared with the Λ-enantiomer. Combined with the previously published research results, experimental results from this study implied that the antitumor activity of a metal complex might have originated from the conformation change of DNA in tumor cells caused by the intercalation of the complex, that the antitumor mechanism of a metal complex could be related to its DNA-binding mode, and that the antitumor efficiency of a metal complex could result from its DNA-binding strength.
Collapse
Affiliation(s)
- Jianrong Jiang
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Qian Chen
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Tianwen Huan
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Yanhong Nie
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Zhongming Dai
- Shenzhen University General Hospital, Shenzhen 518060, China
| | - Dujuan Li
- Key Laboratory of RF Circuits and Systems of Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Xu Xu
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland 1142, New Zealand
| | - Zhangli Hu
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Hong Xu
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Bioresources and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Longhua Innovation Institute for Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
6
|
Raju L, Javan Nikkhah S, K M, Vandichel M, Eswaran R. Anticancer Potential of Dendritic Poly(aryl ether)-Substituted Polypyridyl Ligand-Based Ruthenium(II) Coordination Entities. ACS APPLIED BIO MATERIALS 2023; 6:4226-4239. [PMID: 37782900 DOI: 10.1021/acsabm.3c00452] [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] [Indexed: 10/04/2023]
Abstract
This paper studies the anticancer potency of dendritic poly(aryl ether)-substituted polypyridyl ligand-based ruthenium(II) coordination entities. The dendritic coordination entities were successfully designed, synthesized, and characterized by different spectral methods such as Fourier transform infrared (FTIR), 1H and 13C- NMR, and mass spectrometry. Further, to understand the structure and solvation behavior of the coordination entities, we performed all-atom molecular dynamics (MD) simulations. The behavior, configuration, and size of the coordination entities in DMSO and water were studied by calculating the radius of gyration (Rg) and solvent-accessible surface area (SASA). The MTT assay was used to assess the in vitro cytotoxicity of all of the coordination entities against cancerous A549 (lung cancer cells), MDA MB 231 (breast cancer cells), and HepG2 (liver cancer cells) and was found to be good with comparable IC50 values with respect to the standard drug cisplatin. The coordination entities exhibited dose dependence, and the highest activity was shown against HepG2 cell lines in comparison to the other cancer cell lines. In addition, fluorescence staining studies, such as AO/EB, DAPI, and cell death analysis by PI staining, were performed on the coordination entities to understand the apoptosis mechanism. Furthermore, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) assays confirmed apoptosis in cancer cells via the mitochondrial pathway. The DNA fragmentation assay was done followed by molecular docking analysis with DNA executed to strengthen and support the experimental observations.
Collapse
Affiliation(s)
- Liju Raju
- Department of Chemistry, Madras Christian College (Autonomous), Affiliated to the University of Madras, Tambaram East, Chennai 600059, Tamilnadu, India
| | - Sousa Javan Nikkhah
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick V94 T9PX, Republic of Ireland
| | - MosaChristas K
- Department of Plant Biology and Biotechnology, Loyola Institute of Frontier Energy (LIFE), Loyola College (Autonomous), University of Madras, Chennai 600034, India
| | - Matthias Vandichel
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick V94 T9PX, Republic of Ireland
| | - Rajkumar Eswaran
- Department of Chemistry, Madras Christian College (Autonomous), Affiliated to the University of Madras, Tambaram East, Chennai 600059, Tamilnadu, India
| |
Collapse
|
7
|
Li W, Li S, Xu G, Man X, Yang T, Zhang Z, Liang H, Yang F. Developing a Ruthenium(III) Complex to Trigger Gasdermin E-Mediated Pyroptosis and an Immune Response Based on Decitabine and Liposomes: Targeting Inhibition of Gastric Tumor Growth and Metastasis. J Med Chem 2023; 66:13072-13085. [PMID: 37702429 DOI: 10.1021/acs.jmedchem.3c01110] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
To develop next-generation metal drugs with high efficiency and low toxicity for targeting inhibition of gastric tumor growth and metastasis, we not only optimized a series of ruthenium (Ru, III) 2-hydroxy-1-naphthaldehyde thiosemicarbazone complexes to obtain a Ru(III) complex (4b) with remarkable cytotoxicity in vitro but also constructed a 4b-decitabine (DCT)/liposome (Lip) delivery system (4b-DCT-Lip). The in vivo results showed that 4b-DCT-Lip not only had a stronger capacity to inhibit gastric tumor growth and metastasis than 4b-DCT but also addressed the co-delivery problems of 4b-DCT and improved their targeting ability. Furthermore, we confirmed the mechanism of 4b-DCT/4b-DCT-Lip inhibiting the growth and metastasis of a gastric tumor. DCT-upregulated gasdermin E (GSDME) was cleaved by 4b-activated caspase-3 to afford GSDME-N terminal and then was aggregated to form nonselective pores on the cell membrane of a gastric tumor, thereby inducing pyroptosis and a pyroptosis-induced immune response.
Collapse
Affiliation(s)
- Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Xueyu Man
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Tongfu Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, Guangxi, China
| |
Collapse
|
8
|
Wang J, Zhang Z, Chen Y. Supramolecular immunotherapy on diversiform immune cells. J Mater Chem B 2023; 11:8347-8367. [PMID: 37563947 DOI: 10.1039/d3tb00924f] [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: 08/12/2023]
Abstract
Supramolecular immunotherapy employs supramolecular materials to stimulate the immune system for inhibiting tumor cell growth and metastasis, reducing the cancer recurrence rate, and improving the quality of the patient's life. Additionally, it can lessen patient suffering and the deterioration of their illness, as well as increase their survival rate. This paper will outline the fundamentals of tumor immunotherapy based on supramolecular materials as well as its current state of development and potential applications. To be more specific, we will first introduce the basic principles of supramolecular immunotherapy, including the processes, advantages and limitations of immunotherapy, the construction of supramolecular material structures, and its benefits in treatment. Second, considering the targeting of supramolecular drugs to immune cells, we comprehensively discuss the unique advantages of applying supramolecular drugs with different types of immune cells in tumor immunotherapy. The current research advances in supramolecular immunotherapy, including laboratory research and clinical applications, are also described in detail. Finally, we reveal the tremendous promise of supramolecular materials in tumor immunotherapy, as well as discuss the opportunities and challenges that may be faced in future development.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| | - Ziyi Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, P. R. China
| |
Collapse
|
9
|
Mitchell RJ, Kriger SM, Fenton AD, Havrylyuk D, Pandeya A, Sun Y, Smith T, DeRouchey JE, Unrine JM, Oza V, Blackburn JS, Wei Y, Heidary DK, Glazer EC. A monoadduct generating Ru(ii) complex induces ribosome biogenesis stress and is a molecular mimic of phenanthriplatin. RSC Chem Biol 2023; 4:344-353. [PMID: 37181632 PMCID: PMC10170627 DOI: 10.1039/d2cb00247g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/01/2023] [Indexed: 03/03/2023] Open
Abstract
Ruthenium complexes are often investigated as potential replacements for platinum-based chemotherapeutics in hopes of identifying systems with improved tolerability in vivo and reduced susceptibility to cellular resistance mechanisms. Inspired by phenanthriplatin, a non-traditional platinum agent that contains only one labile ligand, monofunctional ruthenium polypyridyl agents have been developed, but until now, few demonstrated promising anticancer activity. Here we introduce a potent new scaffold, based on [Ru(tpy)(dip)Cl]Cl (tpy = 2,2':6',2''-terpyridine and dip = 4,7-diphenyl-1,10-phenanthroline) in pursuit of effective Ru(ii)-based monofunctional agents. Notably, the extension of the terpyridine at the 4' position with an aromatic ring resulted in a molecule that was cytotoxic in several cancer cell lines with sub-micromolar IC50 values, induced ribosome biogenesis stress, and exhibited minimal zebrafish embryo toxicity. This study demonstrates the successful design of a Ru(ii) agent that mimics many of the biological effects and phenotypes seen with phenanthriplatin, despite numerous differences in both the ligands and metal center structure.
Collapse
Affiliation(s)
- Richard J Mitchell
- Department of Chemistry, University of Kentucky 505 Rose St. Lexington KY 40506 USA
| | - Sarah M Kriger
- Department of Chemistry, North Carolina State University 2620 Yarbrough DriveRaleigh NC 27695 USA
| | - Alexander D Fenton
- Department of Chemistry, University of Kentucky 505 Rose St. Lexington KY 40506 USA
| | - Dmytro Havrylyuk
- Department of Chemistry, University of Kentucky 505 Rose St. Lexington KY 40506 USA
| | - Ankit Pandeya
- Department of Chemistry, University of Kentucky 505 Rose St. Lexington KY 40506 USA
| | - Yang Sun
- Department of Chemistry, University of Kentucky 505 Rose St. Lexington KY 40506 USA
| | - Tami Smith
- Department of Plant and Soil Sciences, University of Kentucky 1100 S. Limestone St Lexington KY 40546 USA
| | - Jason E DeRouchey
- Department of Chemistry, University of Kentucky 505 Rose St. Lexington KY 40506 USA
| | - Jason M Unrine
- Department of Plant and Soil Sciences, University of Kentucky 1100 S. Limestone St Lexington KY 40546 USA
| | - Viral Oza
- Department of Molecular and Cellular Biochemistry, University of Kentucky 741 S. Limestone St. Lexington KY 40536 USA
| | - Jessica S Blackburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky 741 S. Limestone St. Lexington KY 40536 USA
| | - Yinan Wei
- Department of Chemistry, University of Kentucky 505 Rose St. Lexington KY 40506 USA
| | - David K Heidary
- Department of Chemistry, North Carolina State University 2620 Yarbrough DriveRaleigh NC 27695 USA
| | - Edith C Glazer
- Department of Chemistry, North Carolina State University 2620 Yarbrough DriveRaleigh NC 27695 USA
| |
Collapse
|
10
|
Regorafenib and Ruthenium Complex Combination Inhibit Cancer Cell Growth by Targeting PI3K/AKT/ERK Signalling in Colorectal Cancer Cells. Int J Mol Sci 2022; 24:ijms24010686. [PMID: 36614133 PMCID: PMC9820863 DOI: 10.3390/ijms24010686] [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: 10/28/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023] Open
Abstract
Cancer is one of the leading cause of lethality worldwide, CRC being the third most common cancer reported worldwide, with 1.85 million cases and 850,000 deaths annually. As in all other cancers, kinases are one of the major enzymes that play an essential role in the incidence and progression of CRC. Thus, using multi-kinase inhibitors is one of the therapeutic strategies used to counter advanced-stage CRC. Regorafenib is an FDA-approved drug in the third-line therapy of refractory metastatic colorectal cancer. Acquired resistance to cancers and higher toxicity of these drugs are disadvantages to the patients. To counter this, combination therapy is used as a strategy where a minimal dose of drugs can be used to get a higher efficacy and reduce drug resistance development. Ruthenium-based compounds are observed to be a potential alternative to platinum-based drugs due to their significant safety and effectiveness. Formerly, our lab reported Ru-1, a ruthenium-based compound, for its anticancer activity against multiple cancer cells, such as HepG2, HCT116, and MCF7. This study evaluates Ru-1's activity against regorafenib-resistant HCT116 cells and as a combination therapeutic with regorafenib. Meanwhile, the mechanism of the effect of Ru-1 alone and with regorafenib as a combination is still unknown. In this study, we tested a drug combination (Ru-1 and regorafenib) against a panel of HT29, HCT116, and regorafenib-resistant HCT116 cells. The combination showed a synergistic inhibitory activity. Several mechanisms underlying these numerous synergistic activities, such as anti-proliferative efficacy, indicated that the combination exhibited potent cytotoxicity and enhanced apoptosis induction. Disruption of mitochondrial membrane potential increased intracellular ROS levels and decreased migratory cell properties were observed. The combination exhibited its activity by regulating PI3K/Akt and p38 MAP kinase signalling. This indicates that the combination of REG/Ru-1 targets cancer cells by modulating the PI3K/Akt and ERK signalling.
Collapse
|
11
|
Begantsova YE, Baranov EV, Chesnokov SA. 4-(1H-Imidazo[4,5-f][1,10]phenanthrolin-2-yl)benzaldehyde as a probe in pure solvents: Solvatochromism, electric dipole moment and pH influence. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 280:121480. [PMID: 35753103 DOI: 10.1016/j.saa.2022.121480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 06/15/2023]
Abstract
The spectral properties of 4-(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)benzaldehyde (1) in eleven organic solvents of different polarity have been studied. In order to determine the contributions of specific and non-specific interactions between the considered compound and the solvents, the solvatochromic Lippert-Mataga, McRae, Bakhshiev methods have been applied. The compound demonstrates positive solvatochromism. The dipole moment of the excited state of 1 obtained using the Reichardt method is equal to 10.56/7.08 D for trans- and cis-conformers, respectively, and agrees well with the theoretically calculated value. The influence of the polarizability of 1 on changes in the dipole moments has been analyzed using the Bilot-Kawski method. The multiple linear regression analysis in the framework of the Kamlet-Abboud-Taft and Catalán models has highlighted that the main properties which determine the Stokes shift of 1 are the acidity and dipolarity of the solvent. The variation of pH by additions of acid or base to solution 1 leads to significant changes in absorption and fluorescence spectra, therefore, 1 can be of interest as a solvatochromic probe, being sensitive to acidic/base properties of the environment. It has also been found out that the anion form of 1 is present in the DMSO solution. An addition of N,N-dimethylcyclohexylamine intensifies the dissociation of the considered compound in the DMSO solution and suppresses the fluorescence at a large amine excess.
Collapse
Affiliation(s)
- Yulia E Begantsova
- G.A. Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, 49 Tropinin Str., Nizhny Novgorod 603950, Russian Federation.
| | - Evgeny V Baranov
- G.A. Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, 49 Tropinin Str., Nizhny Novgorod 603950, Russian Federation
| | - Sergey A Chesnokov
- G.A. Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, 49 Tropinin Str., Nizhny Novgorod 603950, Russian Federation
| |
Collapse
|
12
|
Ma X, Lu J, Yang P, Zhang Z, Huang B, Li R, Ye R. 8-Hydroxyquinoline-modified ruthenium(II) polypyridyl complexes for JMJD inhibition and photodynamic antitumor therapy. Dalton Trans 2022; 51:13902-13909. [PMID: 36040403 DOI: 10.1039/d2dt01765b] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
As an ideal scaffold for metal ion chelation, 8-hydroxyquinoline (8HQ) can chelate different metal ions, such as Fe2+, Cu2+, Zn2+, etc. Here, by integrating 8HQ with a ruthenium(II) polypyridyl moiety, two Ru(II)-8HQ complexes (Ru1 and Ru2), [Ru(N-N)2L](PF6)2 (L = 2-(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)quinolin-8-ol; N-N: 2,2'-bipyridine (bpy, in Ru1), 1,10-phenanthroline (phen, in Ru2)) were designed and synthesized. In both complexes, ligand L is an 8HQ derivative designed to chelate the cofactor Fe2+ of jumonji C domain-containing demethylase (JMJD). As expected, Ru1 and Ru2 could inhibit the activity of JMJD by chelating the key cofactor Fe2+ of JMJD, resulting in the upregulation of histone-methylation levels in human lung cancer (A549) cells, and the upregulation was more pronounced under light conditions. In addition, MTT data showed that Ru1 and Ru2 exhibited lower dark toxicity, and light irradiation could significantly enhance their antitumor activity. The marked photodynamic activities of Ru1 and Ru2 could induce the elevation of reactive oxygen species (ROS), depolarization of mitochondrial membrane potential (MMP), and activation of caspases. These mechanistic studies indicated that Ru1 and Ru2 could induce apoptosis through the combination of JMJD inhibitory and PDT activities, thereby achieving dual antitumor effects.
Collapse
Affiliation(s)
- Xiurong Ma
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Junjian Lu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Peixin Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Zheng Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Bo Huang
- Faculty of Chemistry and Chemical Engineering, Yunnan Normal University, Kunming 650500, P. R. China.
| | - Rongtao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Ruirong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| |
Collapse
|
13
|
Peng F, Liao M, Qin R, Zhu S, Peng C, Fu L, Chen Y, Han B. Regulated cell death (RCD) in cancer: key pathways and targeted therapies. Signal Transduct Target Ther 2022; 7:286. [PMID: 35963853 PMCID: PMC9376115 DOI: 10.1038/s41392-022-01110-y] [Citation(s) in RCA: 302] [Impact Index Per Article: 151.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023] Open
Abstract
Regulated cell death (RCD), also well-known as programmed cell death (PCD), refers to the form of cell death that can be regulated by a variety of biomacromolecules, which is distinctive from accidental cell death (ACD). Accumulating evidence has revealed that RCD subroutines are the key features of tumorigenesis, which may ultimately lead to the establishment of different potential therapeutic strategies. Hitherto, targeting the subroutines of RCD with pharmacological small-molecule compounds has been emerging as a promising therapeutic avenue, which has rapidly progressed in many types of human cancers. Thus, in this review, we focus on summarizing not only the key apoptotic and autophagy-dependent cell death signaling pathways, but the crucial pathways of other RCD subroutines, including necroptosis, pyroptosis, ferroptosis, parthanatos, entosis, NETosis and lysosome-dependent cell death (LCD) in cancer. Moreover, we further discuss the current situation of several small-molecule compounds targeting the different RCD subroutines to improve cancer treatment, such as single-target, dual or multiple-target small-molecule compounds, drug combinations, and some new emerging therapeutic strategies that would together shed new light on future directions to attack cancer cell vulnerabilities with small-molecule drugs targeting RCD for therapeutic purposes.
Collapse
Affiliation(s)
- Fu Peng
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minru Liao
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shiou Zhu
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Leilei Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Yi Chen
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
14
|
Shahzad K, Asad M, Asiri AM, Irfan M, Iqbal MA. In-vitro anticancer profile of recent ruthenium complexes against liver cancer. REV INORG CHEM 2022. [DOI: 10.1515/revic-2021-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Ruthenium complexes are considered as the most favorable alternatives to traditional platinum-based cancer drugs owing to their acceptable toxicity level, selectivity, variant oxidation states and ability to treat platinum-resistant cancer cells. They have similar ligand exchange kinetics as platinum drugs but can be tailored according to our desire by ligands influence. In the current study, we illustrate the in-vitro anticancer profile of some ruthenium complexes (2016–2021) against human hepatocellular carcinoma (HepG2). The anticancer activity of ruthenium complexes is determined by comparing their IC50 values with one another and positive controls. Fortunately, some ruthenium complexes including 3, 4, 6, 14, 15, 20, 42, and 48 exhibit surpassed in-vitro anticancer profile than that of positive controls promising as potential candidates against liver cancer. We also explored the structure-activity relationship (SAR) which is a key factor in the rational designing and synthesis of new ruthenium drugs. It covers the factors affecting anticancer activity including lipophilicity, planarity, area and bulkiness, the steric influence of different ligands, and electronic effects induced by ligands, stability, aqueous solubility and bioavailability to the target sites. The data reported here will provide strong support in the plausible design and synthesis of ruthenium anticancer drugs in the upcoming days.
Collapse
Affiliation(s)
- Khurram Shahzad
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
| | - Mohammad Asad
- Center of Excellence for Advanced Materials Research (CEAMR) , King Abdulaziz University , P.O. Box 80203 , Jeddah 21589 , Saudi Arabia
- Chemistry Department , Faculty of Science, King Abdulaziz University , P.O. Box 80203 , Jeddah 21589 , Saudi Arabia
| | - Abdullah M. Asiri
- Center of Excellence for Advanced Materials Research (CEAMR) , King Abdulaziz University , P.O. Box 80203 , Jeddah 21589 , Saudi Arabia
- Chemistry Department , Faculty of Science, King Abdulaziz University , P.O. Box 80203 , Jeddah 21589 , Saudi Arabia
| | - Muhammad Irfan
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
| | - Muhammad Adnan Iqbal
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
- Organometallic and Coordination Chemistry Laboratory , University of Agriculture , Faisalabad , 38000 , Pakistan
| |
Collapse
|
15
|
Ruthenium complexes boost NK cell immunotherapy via sensitizing triple-negative breast cancer and shaping immuno-microenvironment. Biomaterials 2022; 281:121371. [DOI: 10.1016/j.biomaterials.2022.121371] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 01/03/2022] [Accepted: 01/08/2022] [Indexed: 11/23/2022]
|
16
|
Antimicrobial effect of Casiopeinas® copper- and ruthenium-based compounds on Aggregatibacter actinomycetemcomitans and in vitro cell viability onto osteoblasts cells. Braz J Microbiol 2021; 53:179-184. [PMID: 34741282 DOI: 10.1007/s42770-021-00648-3] [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: 03/09/2021] [Accepted: 10/26/2021] [Indexed: 10/19/2022] Open
Abstract
OBJECTIVES The present study aims to evaluate the antimicrobial property of Casiopeinas® copper- and ruthenium-based compounds against Aggregatibacter actinomycetemcomitans serotype b (ATCC® 43,718™), as well as the cytotoxicity on an osteoblasts cell line of both compounds. MATERIAL AND METHODS The antibacterial effect of the copper-based compounds (CasII-gly, CasIII-ia) and the ruthenium-based compound (RuN-6) at four different concentrations was evaluated as the inhibition ratio of the bacterial growth after 48 h under anaerobic conditions, and the cell viability was measured through resazurin assay. RESULTS The copper- and ruthenium-based compounds used for this assay were (CasII-gly, CasIII-ia, and RuN-6), showing inhibitory activity between 39 and 62% compared to the antibiotic employed as control 66%. Cell viability was established between 61 and 96%. CONCLUSIONS Casiopeinas® and ruthenium showed dose and time dependent, inhibitory activity on A. actinomycetemcomitans, and low toxicity on cells (osteoblast) underexposure. The compound CasII-gly showed the best antimicrobial effect, and it could be considered a possible antimicrobial agent in periodontal therapy.
Collapse
|
17
|
Zhou Y, Bai L, Tian L, Yang L, Zhang H, Zhang Y, Hao J, Gu Y, Liu Y. Iridium(III)-BBIP complexes induce apoptosis via PI3K/AKT/mTOR pathway and inhibit A549 lung tumor growth in vivo. J Inorg Biochem 2021; 223:111550. [PMID: 34311319 DOI: 10.1016/j.jinorgbio.2021.111550] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
The new ligand BBIP (BBIP = 2-(7-bromo-2H-benzo[d]imidazole-4-yl)-1H-imidazo[4,5-f][1,10]phenanthroline) with its iridium(III) complexes: [Ir(ppy)2(BBIP)](PF6) (ppy = 2-phenylpyridine, Ir1), [Ir(bzq)2(BBIP)](PF6) (bzq = benzo[h]quinolone, Ir2) and [Ir(piq)2(BBIP)](PF6) (piq = 1-phenylisoquinoline, Ir3) were synthesized and characterized by elemental analysis, High Resolution Mass Spectrometer (HRMS), 1H NMR and 13C{1H} NMR. The cytotoxicity of the complexes against A549, HepG2, SGC-7901, BEL-7402, HeLa and normal LO2 was evaluated through 3-(4,5-dimethylthiazole-2-yl)-2,5-biphenyl tetrazolium bromide (MTT) method. The results show that Ir1 exhibits high cytotoxic activity against A549 cells with a low IC50 value of 4.9 ± 0.5 μM. A series of biological activities such as cell cycle arrest, endoplasmic reticulum localization assay, apoptosis, western blotting, cellular uptake determination and in vivo antitumor activity were investigated. The assays implied that the complexes inhibit cancer cell migration through blocking mitotic progress. Cell cycle distribution stated that the complexes depress cell growth at G0/G1 phase. Additionally, the complexes acted on the endoplasmic reticulum and induce apoptosis through endoplasmic reticulum stress pathway. Especially, the western blotting showed that the complexes activated Bcl-2 (B-cell lymphoma-2) family and decreased PI3K (phosphoinositide-3 kinase) and AKT (protein kinase B), up-regulated the expression of mTOR (mammalian target of rapamycin) and p-mTOR (phosphorylated mammalian target of rapamycin). Therefore, the complexes induce apoptosis through activating PI3K-AKT-mTOR pathway. Antitumor in vivo demonstrated that Ir1 can effectively prevent the tumor growth with an inhibitory rate of 48.89%.
Collapse
Affiliation(s)
- Yi Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lan Bai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Li Tian
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Linlin Yang
- Department of Pediatrics, Guangdong Women and Children Hospital, Guangzhou 510010, PR China.
| | - Huiwen Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yuanyuan Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jing Hao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yiying Gu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
18
|
Yang M, Zhao H, Zhang Z, Yuan Q, Feng Q, Duan X, Wang S, Tang Y. CO/light dual-activatable Ru(ii)-conjugated oligomer agent for lysosome-targeted multimodal cancer therapeutics. Chem Sci 2021; 12:11515-11524. [PMID: 34667555 PMCID: PMC8447874 DOI: 10.1039/d1sc01317c] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/21/2021] [Indexed: 01/10/2023] Open
Abstract
Stimuli-activatable and subcellular organelle-targeted agents with multimodal therapeutics are urgently desired for highly precise and effective cancer treatment. Herein, a CO/light dual-activatable Ru(ii)-oligo-(thiophene ethynylene) (Ru-OTE) for lysosome-targeted cancer therapy is reported. Ru-OTE is prepared via the coordination-driven self-assembly of a cationic conjugated oligomer (OTE-BN) ligand and a Ru(ii) center. Upon the dual-triggering of internal gaseous signaling molecular CO and external light, Ru-OTE undergoes ligand substitution and releases OTE-BN followed by dramatic fluorescence recovery, which could be used for monitoring drug delivery and imaging guided anticancer treatments. The released OTE-BN selectively accumulates in lysosomes, physically breaking their integrity. Then, the generated cytotoxic singlet oxygen (1O2) causes severe lysosome damage, thus leading to cancer cell death via photodynamic therapy (PDT). Meanwhile, the release of the Ru(ii) core also suppresses cancer cell growth as an anticancer metal drug. Its significant anticancer effect is realized via the multimodal therapeutics of physical disruption/PDT/chemotherapy. Importantly, Ru-OTE can be directly photo-activated using a two-photon laser (800 nm) for efficient drug release and near-infrared PDT. Furthermore, Ru-OTE with light irradiation inhibits tumor growth in an MDA-MB-231 breast tumor model with negligible side effects. This study demonstrates that the development of an activatable Ru(ii)-conjugated oligomer potential drug provides a new strategy for effective subcellular organelle-targeted multimodal cancer therapeutics. The anticancer therapeutics of lysosome disruption/PDT/chemotherapy based on Ru-OTE complex was achieved, which provides a new strategy for developing multimodal and effective stimuli-activatable subcellular organelle-targeted cancer therapeutics.![]()
Collapse
Affiliation(s)
- Min Yang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Hao Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences P. R. China
| | - Ziqi Zhang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Qiong Yuan
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Qian Feng
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Xinrui Duan
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences P. R. China
| | - Yanli Tang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University Xi'an Shaanxi Province 710119 P. R. China
| |
Collapse
|
19
|
Rinaldi-Neto F, Ribeiro AB, Ferreira NH, Squarisi IS, Oliveira KM, Orenha RP, Parreira RLT, Batista AA, Tavares DC. Anti-melanoma effect of ruthenium(II)-diphosphine complexes containing naphthoquinone ligand. J Inorg Biochem 2021; 222:111497. [PMID: 34090039 DOI: 10.1016/j.jinorgbio.2021.111497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022]
Abstract
The use of natural products as potential ligands has been explored as a strategy in the development of metal-based chemotherapy. Since ruthenium complexes are promising alternatives to traditional antitumor agents, this study evaluated the anti-melanoma potential of two ruthenium(II) complexes containing the naphthoquinone ligands lapachol (lap), [Ru(lap)(dppm)2]PF6, and lawsone (law), [Ru(law)(dppm)2]PF6, in addition to the bis(diphenylphosphino)methane (dppm) ligand, referred to as complexes (1) and (2), respectively, using a syngeneic murine melanoma model. Activation of the apoptotic pathway by the treatments was assessed by immunohistochemistry in tumor tissue. Additionally, toxicity of the treatments was evaluated by variation in body and organ weight, quantification of biochemical indicators of renal damage, and genotoxicity in bone marrow and hepatocytes. First, the antiproliferative activity of (1) and (2) was observed in B16F10 cells, with IC50 values of 2.78 and 1.68 μM, respectively. The results obtained in mice showed that, unlike complex (1), (2) possesses significant anti-melanoma activity demonstrated by a reduction in tumor volume and mass (88.42%), as well as in mitosis frequency (83.86%). Additionally, complex (2) increased the levels of cleaved caspase-3, inducing tumor cell apoptosis. When compared to the metallodrug cisplatin, complex (2) exhibited similar anti-melanoma activity and lower toxicity considering all parameters evaluated. In silico studies demonstrated no difference in the binding energy of the naphthoquinone complex between complexes (1) and (2). However, the complex containing the lawsone ligand has a lower molar volume, which may be important for interactions with minor DNA grooves. The present results demonstrate the antitumor efficiency of complex (2) and a significantly lower systemic toxicity compared to cisplatin.
Collapse
Affiliation(s)
- Francisco Rinaldi-Neto
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Arthur Barcelos Ribeiro
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Natália Helen Ferreira
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Iara Silva Squarisi
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Kátia Mara Oliveira
- Universidade Federal de São Carlos, Departamento de Química, Rodovia Washington Luis s/n Km 235, São Carlos, São Paulo 13565-905, Brazil
| | - Renato Pereira Orenha
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Renato Luís Tame Parreira
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Alzir Azevedo Batista
- Universidade Federal de São Carlos, Departamento de Química, Rodovia Washington Luis s/n Km 235, São Carlos, São Paulo 13565-905, Brazil
| | - Denise Crispim Tavares
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, 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
|
Chen M, Huang X, Shi H, Lai J, Ma L, Lau TC, Chen T. Cr(V)-Cr(III) in-situ transition promotes ROS generation to achieve efficient cancer therapy. Biomaterials 2021; 276:120991. [PMID: 34237506 DOI: 10.1016/j.biomaterials.2021.120991] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/19/2021] [Accepted: 06/22/2021] [Indexed: 01/29/2023]
Abstract
The development of metal-based anticancer drugs is of considerable interest and significance in inorganic medicine. In contrast to noble metal-based small molecules, the anticancer property of earth abundant metal-based small molecules is much less explored which are usually essential trace element for the human body. Among earth abundant metals, chromium (Cr) in the +3 valent is an essential trace element for the human body to low down the blood lipids and maintain the blood sugar; on the other hand, Cr(VI) are known to be highly toxic due to their oxidation power. To design stable high-valent Cr small molecules to construct Cr(high-valent)-Cr(III) in-situ transition system to achieve low-toxic and highly efficient anti-cancer therapy is a very desirable approach. Herein we report the Cr(V)-Cr(III) in-situ transition system promotes ROS generation to achieve efficient cancer therapy in vivo and in vitro. To the best of our knowledge, these Cr-based small molecules are the first stable Cr(V) compounds with potent anticancer efficacy, especially towards malignant cancers.
Collapse
Affiliation(s)
- Mingkai Chen
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China
| | - Xiaoting Huang
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China
| | - Huatian Shi
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong, China
| | - Jie Lai
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China
| | - Li Ma
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Tai-Chu Lau
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong, China.
| | - Tianfeng Chen
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
22
|
Hsieh MC, Lo YS, Chuang YC, Lin CC, Ho HY, Hsieh MJ, Lin JT. Dehydrocrenatidine extracted from Picrasma quassioides induces the apoptosis of nasopharyngeal carcinoma cells through the JNK and ERK signaling pathways. Oncol Rep 2021; 46:166. [PMID: 34165177 PMCID: PMC8218301 DOI: 10.3892/or.2021.8117] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an indicator disease in Asia due to its unique geographical and ethnic distribution. Dehydrocrenatidine (DC) is a β-carboline alkaloid abundantly present in Picrasma quassioides (D. Don) Benn, a deciduous shrub or small tree native to temperate regions of southern Asia, and β-carboline alkaloids play anti-inflammatory and antiproliferative roles in various cancers. However, the mechanism and function of DC in human NPC cells remain only partially explored. The present study aimed to examine the cytotoxicity and biochemical role of DC in human NPC cells. The MTT method, cell cycle analysis, DAPI determination, Annexin V/PI double staining, and mitochondrial membrane potential examination were performed to evaluate the effects of DC treatment on human NPC cell lines. In addition, western blotting analysis was used to explore the effect of DC on apoptosis and signaling pathways in related proteins. The analysis results confirmed that DC significantly reduced the viability of NPC cell lines in a dose- and time-dependent manner and induced apoptosis through internal and external apoptotic pathways (including cell cycle arrest, altered mitochondrial membrane potential, and activated death receptors). Western blot analysis illustrated that DC's effect on related proteins in the mitogen-activated protein kinase pathway can induce apoptosis by enhancing ERK phosphorylation and inhibiting Janus kinase (JNK) phosphorylation. Notably, DC induced apoptosis by affecting the phosphorylation of JNK and ERK, and DC and inhibitors (SP600125 and U0126) in combination restored the overexpression of p-JNK and p-ERK. To date, this is the first study to confirm the apoptosis pathway induced by DC phosphorylation of p-JNK and p-REK in human NPC. On the basis of evidence obtained from this study, DC targeting the inhibition of NPC cell lines may be a promising future strategy for NPC treatment.
Collapse
Affiliation(s)
- Ming-Chang Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan, R.O.C
| | - Yu-Sheng Lo
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan, R.O.C
| | - Yi-Ching Chuang
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan, R.O.C
| | - Chia-Chieh Lin
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan, R.O.C
| | - Hsin-Yu Ho
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan, R.O.C
| | - Ming-Ju Hsieh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan, R.O.C
| | - Jen-Tsun Lin
- Post Baccalaureate Medicine, National Chung Hsing University, Taichung 402, Taiwan, R.O.C
| |
Collapse
|
23
|
BEZ235 Increases the Sensitivity of Hepatocellular Carcinoma to Sorafenib by Inhibiting PI3K/AKT/mTOR and Inducing Autophagy. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5556306. [PMID: 33987439 PMCID: PMC8079203 DOI: 10.1155/2021/5556306] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/13/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022]
Abstract
Acquired resistance of hepatocellular carcinoma (HCC) to sorafenib (SFB) is the main reason for the failure of SFB treatment of the cancer. Abnormal activation of the PI3K/AKT/mTOR pathway is important in the acquired resistance of SFB. Therefore, we investigated whether BEZ235 (BEZ) could reverse acquired sorafenib resistance by targeting the PI3K/mTOR pathway. A sorafenib-resistant HCC cell line Huh7R was established. MTT assay, clone formation assay, flow cytometry, and immunofluorescence were used to analyze the effects of BEZ235 alone or combined with sorafenib on cell proliferation, cell cycle, apoptosis, and autophagy of Huh7 and Huh7R cells. The antitumor effect was evaluated in animal models of Huh7R xenografts in vivo. Western blot was used to detect protein levels of the PI3K/AKT/mTOR pathway and related effector molecules. In vitro results showed that the Huh7R had a stronger proliferation ability and antiapoptosis effect than did Huh7, and sorafenib had no inhibitory effect on Huh7R. SFB + BEZ inhibited the activation of the PI3K/AKT/mTOR pathway caused by sorafenib. Moreover, SFB + BEZ inhibited the proliferation and cloning ability, blocked the cell cycle in the G0/G1 phase, and promoted apoptosis in the two cell lines. The autophagy level in Huh7R cells was higher than in Huh7 cells, and BEZ or SFB + BEZ further promoted autophagy in the two cell lines. In vivo, SFB + BEZ inhibited tumor growth by inducing apoptosis and autophagy. We concluded that BEZ235 enhanced the sensitivity of sorafenib through suppressing the PI3K/AKT/mTOR pathway and inducing autophagy. These observations may provide the experimental basis for sorafenib combined with BEZ235 in trial treatment of HCC.
Collapse
|
24
|
Gul NS, Khan TM, Chen M, Huang KB, Hou C, Choudhary MI, Liang H, Chen ZF. New copper complexes inducing bimodal death through apoptosis and autophagy in A549 cancer cells. J Inorg Biochem 2020; 213:111260. [DOI: 10.1016/j.jinorgbio.2020.111260] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/13/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022]
|
25
|
Synthesis and anti-cancer activity of bis-amino-phosphine ligand and its ruthenium(II) complexes. Bioorg Med Chem Lett 2020; 30:127492. [DOI: 10.1016/j.bmcl.2020.127492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 08/06/2020] [Accepted: 08/09/2020] [Indexed: 12/30/2022]
|
26
|
Costa MS, Gonçalves YG, Borges BC, Silva MJB, Amstalden MK, Costa TR, Antunes LMG, Rodrigues RS, Rodrigues VDM, de Faria Franca E, Zoia MAP, de Araújo TG, Goulart LR, Von Poelhsitz G, Yoneyama KAG. Ruthenium (II) complex cis-[Ru II(ŋ 2-O 2CC 7H 7O 2)(dppm) 2]PF 6-hmxbato induces ROS-mediated apoptosis in lung tumor cells producing selective cytotoxicity. Sci Rep 2020; 10:15410. [PMID: 32958783 PMCID: PMC7506019 DOI: 10.1038/s41598-020-72420-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/31/2020] [Indexed: 12/24/2022] Open
Abstract
Ruthenium complexes have been extensively explored as potential molecules for cancer treatment. Considering our previous findings on the remarkable cytotoxic activity exhibited by the ruthenium (II) complex 3-hydroxy-4-methoxybenzoate (hmxbato)-cis-[RuII(ŋ2-O2CC7H7O2)(dppm)2]PF6 against Leishmania promastigotes and also the similar metabolic characteristics between trypanosomatids and tumor cells, the present study aimed to analyze the anticancer potential of hmxbato against lung tumor cells, as well as the partial death mechanisms involved. Hmxbato demonstrated selective cytotoxicity against A549 lung tumor cells. In addition, this complex at a concentration of 3.8 µM was able to expressively increase the generation of reactive oxygen species (ROS) in tumor cells, causing an oxidative stress that may culminate in: (1) reduction in cellular proliferation; (2) changes in cell morphology and organization patterns of the actin cytoskeleton; (3) cell arrest in the G2/M phase of the cell cycle; (4) apoptosis; (5) changes in the mitochondrial membrane potential and (6) initial DNA damage. Furthermore, we demonstrated that the induction of programmed cell death can occur by the intrinsic apoptotic pathway through the activation of caspases. It is also worth highlighting that hmxbato exhibited predominant actions on A549 tumor cells in comparison to BEAS-2B normal bronchial epithelium cells, which makes this complex an interesting candidate for the design of new drugs against lung cancer.
Collapse
Affiliation(s)
- Mônica Soares Costa
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil.
| | | | - Bruna Cristina Borges
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Marcelo José Barbosa Silva
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Martin Krähenbühl Amstalden
- Departamento de Análises Clínicas, Toxicologia e Ciências Alimentares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, CEP 14040-903, Brazil
| | - Tássia Rafaella Costa
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil
| | - Lusânia Maria Greggi Antunes
- Departamento de Análises Clínicas, Toxicologia e Ciências Alimentares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, CEP 14040-903, Brazil
| | - Renata Santos Rodrigues
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil
| | - Veridiana de Melo Rodrigues
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil
| | - Eduardo de Faria Franca
- Laboratório de Cristalografia e Química Computacional, Instituto de Química, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Mariana Alves Pereira Zoia
- Laboratório de Nanobiotecnologia, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Thaise Gonçalves de Araújo
- Laboratório de Nanobiotecnologia, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Luiz Ricardo Goulart
- Laboratório de Nanobiotecnologia, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Gustavo Von Poelhsitz
- Instituto de Química, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Kelly Aparecida Geraldo Yoneyama
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil.
| |
Collapse
|
27
|
Li J, Chen T. Transition metal complexes as photosensitizers for integrated cancer theranostic applications. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213355] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
28
|
Synthesis, evaluation of biological activity studies of iridium(III) complexes against human gastric carcinoma SGC-7901 cells. INORG CHEM COMMUN 2020. [DOI: 10.1016/j.inoche.2020.108012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
29
|
Ferraro MG, Piccolo M, Misso G, Maione F, Montesarchio D, Caraglia M, Paduano L, Santamaria R, Irace C. Breast Cancer Chemotherapeutic Options: A General Overview on the Preclinical Validation of a Multi-Target Ruthenium(III) Complex Lodged in Nucleolipid Nanosystems. Cells 2020; 9:E1412. [PMID: 32517101 PMCID: PMC7349411 DOI: 10.3390/cells9061412] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
In this review we have showcased the preclinical development of original amphiphilic nanomaterials designed for ruthenium-based anticancer treatments, to be placed within the current metallodrugs approach leading over the past decade to advanced multitarget agents endowed with limited toxicity and resistance. This strategy could allow for new options for breast cancer (BC) interventions, including the triple-negative subtype (TNBC) with poor therapeutic alternatives. BC is currently the second most widespread cancer and the primary cause of cancer death in women. Hence, the availability of novel chemotherapeutic weapons is a basic requirement to fight BC subtypes. Anticancer drugs based on ruthenium are among the most explored and advanced next-generation metallotherapeutics, with NAMI-A and KP1019 as two iconic ruthenium complexes having undergone clinical trials. In addition, many nanomaterial Ru complexes have been recently conceived and developed into anticancer drugs demonstrating attractive properties. In this field, we focused on the evaluation of a Ru(III) complex-named AziRu-incorporated into a suite of both zwitterionic and cationic nucleolipid nanosystems, which proved to be very effective for the in vivo targeting of breast cancer cells (BBC). Mechanisms of action have been widely explored in the context of preclinical evaluations in vitro, highlighting a multitarget action on cell death pathways which are typically deregulated in neoplasms onset and progression. Moreover, being AziRu inspired by the well-known NAMI-A complex, information on non-nanostructured Ru-based anticancer agents have been included in a precise manner.
Collapse
Affiliation(s)
- Maria Grazia Ferraro
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Gabriella Misso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (G.M.); (M.C.)
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 421, 80126 Naples, Italy; (D.M.); (L.P.)
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (G.M.); (M.C.)
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 421, 80126 Naples, Italy; (D.M.); (L.P.)
| | - Rita Santamaria
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| |
Collapse
|
30
|
Zhou YJ, Chen JM, Sapkota K, Long JY, Liao YJ, Jiang JJ, Liang BY, Wei JB, Zhou Y. Pananx notoginseng saponins attenuate CCL2-induced cognitive deficits in rats via anti-inflammation and anti-apoptosis effects that involve suppressing over-activation of NMDA receptors. Biomed Pharmacother 2020; 127:110139. [PMID: 32302948 DOI: 10.1016/j.biopha.2020.110139] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/22/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
HIV-associated neurocognitive disorders (HAND) are characterized by synaptic damage and neuronal loss in the brain, ultimately leading to progressive decline of cognitive abilities and memory. Chemokine CC motif ligand 2 (CCL2) is elevated in cerebrospinal fluid (CSF), and has been believed to contribute to HAND. Previous studies by our research team have shown that CCL2 enhances N-Methyl-D-aspartate receptor (NMDAR)-mediated excitatory postsynaptic currents (EPSCs) and causes nerve cell damage. However, there are few drugs currently available to treat nerve damage that is caused by CCL2. Panax notoginseng saponins (PNS) are isolated from Panax notoginseng and benefit the human body in various ways, including the neuroprotective effect. However, the protective effect of PNS on CCL2-induced neurotoxicity remains unknown. In this study, we found that PNS improved CCL2-induced learning and memory impairment, and inhibited CCL2-induced cell death. These effects may be due to inhibiting over-activation of NMDA receptors by alleviating the dysfunction of glutamate metabolism. Furthermore, PNS-modulated CCL2-inducd intracellular oxidative stress was found to attenuate cell inflammation. Additionally, PNS pretreatment evidently inhibited apoptotic pathways by reducing the Bax/BCL-2 ratio and caspase-3, 8, 9 expressions. In conclusion, this study demonstrates that PNS provides substantial neuroprotection against CCL2-induced neurotoxicity, and may be a novel therapeutic agent in CCL2-induced HAND or other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Jun Zhou
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Jian-Min Chen
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Kiran Sapkota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, United States
| | - Jiang-Yi Long
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yuan-Jun Liao
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jun-Jun Jiang
- Guangxi key laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Bing-Yu Liang
- Guangxi key laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jin-Bin Wei
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Yan Zhou
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
31
|
Lin K, Rong Y, Chen D, Zhao Z, Bo H, Qiao A, Hao X, Wang J. Combination of Ruthenium Complex and Doxorubicin Synergistically Inhibits Cancer Cell Growth by Down-Regulating PI3K/AKT Signaling Pathway. Front Oncol 2020; 10:141. [PMID: 32133289 PMCID: PMC7041628 DOI: 10.3389/fonc.2020.00141] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/27/2020] [Indexed: 01/21/2023] Open
Abstract
Combinational use of drugs has been a common strategy in cancer treatment because of synergistic advantages in reducing dose and toxicity, minimizing or delaying drug resistance. To improve the efficacy of chemotherapy, various potential combinations have been investigated. Ruthenium complex is considered a potential alternative of the platinum-based drugs due to its significant efficacy and safety. Previously, we reported that ruthenium(II) complex (Δ-Ru1) has great anticancer potential and minor toxicity toward normal tissues. However, the therapeutic efficacy and mechanism of action of ruthenium(II) complex combined with other anticancer drugs is still unknown. Here, we investigated the combinational effect of Δ-Ru1 and doxorubicin in different cancer cells. The data assessed by Chou-Talalay method showed significant synergism in MCF-7 cells. Furthermore, the results in antiproliferation efficacy indicated that the combination showed strong cytotoxicity and increasing apoptosis of MCF-7 cells in 2D and 3D multicellular tumor spheroids (MCTSs). Significant inhibition of MCF-7 cells accompanied with increased ROS generation was observed. Furthermore, the expression of PI3K/AKT was significantly down-regulated, while the expression of PTEN was strongly up-regulated in cells treated with combination of Δ-Ru1 and doxorubicin. The expression of NF-κB and XIAP decreased while the expression of P53 increased and associated with apoptosis. These findings suggest that the combination of ruthenium complex and doxorubicin has a significant synergistic effect by down-regulating the PI3K/AKT signaling pathway in MCF-7 cells. This study may trigger more research in ruthenium complex and combination therapy that will be able to provide opportunities for developing better therapeutics for cancer treatment.
Collapse
Affiliation(s)
- Ke Lin
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Rong
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dan Chen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zizhuo Zhao
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huaben Bo
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Aimin Qiao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaojuan Hao
- Manufacturing, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
| | - Jinquan Wang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
32
|
|
33
|
Liu J, Lai H, Xiong Z, Chen B, Chen T. Functionalization and cancer-targeting design of ruthenium complexes for precise cancer therapy. Chem Commun (Camb) 2019; 55:9904-9914. [PMID: 31360938 DOI: 10.1039/c9cc04098f] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The successful clinical application of the three generation platinum anticancer drugs, cisplatin, carboplatin and oxaliplatin, has promoted research interest in metallodrugs; however, the problems of drug resistance and adverse effects have hindered their further application and effects. Thus, scientists are searching for new anticancer metallodrugs with lower toxicity and higher efficacy. The ruthenium complexes have emerged as the most promising alternatives to platinum-based anticancer agents because of their unique multifunctional biochemical properties. In this review, we first focus on the anticancer applications of various ruthenium complexes in different signaling pathways, including the mitochondria-mediated pathway, the DNA damage-mediated pathway, and the death receptor-mediated pathway. We then discuss the functionalization and cancer-targeting designs of different ruthenium complexes in conjunction with other therapies such as photodynamic therapy, photothermal therapy, radiosensitization, targeted therapy and nanotechnology for precise cancer therapy. This review will help in designing and accelerating the research progress regarding new anticancer ruthenium complexes.
Collapse
Affiliation(s)
- Jinggong Liu
- Orthopedics Department, Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou 510120, China
| | | | | | | | | |
Collapse
|
34
|
Lai H, Zeng D, Liu C, Zhang Q, Wang X, Chen T. Selenium-containing ruthenium complex synergizes with natural killer cells to enhance immunotherapy against prostate cancer via activating TRAIL/FasL signaling. Biomaterials 2019; 219:119377. [DOI: 10.1016/j.biomaterials.2019.119377] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022]
|
35
|
Liu XW, Huang J, Tang YX, Zhang SB, Lu JL. Topoisomerase I inhibitory and photocleavage activity by ruthenium complexes containing a new polyaza ligand. INORG NANO-MET CHEM 2019. [DOI: 10.1080/24701556.2019.1661446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Xue-Wen Liu
- College of Chemistry and Material Engineering, Hunan University of Arts and Science, Chang De, P.R. China
| | - Jie Huang
- College of Chemistry and Material Engineering, Hunan University of Arts and Science, Chang De, P.R. China
| | - Yu-Xuan Tang
- College of Chemistry and Material Engineering, Hunan University of Arts and Science, Chang De, P.R. China
| | - Song-Bai Zhang
- College of Chemistry and Material Engineering, Hunan University of Arts and Science, Chang De, P.R. China
| | - Ji-Lin Lu
- College of Chemistry and Material Engineering, Hunan University of Arts and Science, Chang De, P.R. China
| |
Collapse
|
36
|
Haghdoost MM, Golbaghi G, Guard J, Sielanczyk S, Patten SA, Castonguay A. Synthesis, characterization and biological evaluation of cationic organoruthenium(ii) fluorene complexes: influence of the nature of the counteranion. Dalton Trans 2019; 48:13396-13405. [PMID: 31432885 DOI: 10.1039/c9dt00143c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this study, five ruthenium arene complexes with fluorene-bearing N,N-(1) and N,O-(2) donor Schiff base ligands were synthesized and fully characterized. Cationic ruthenium complexes 3[X], ([Ru(η6-C6H6)(Cl)(fluorene-N[double bond, length as m-dash]CH-pyridine)][X] (where X = BF4, PF6, BPh4), were obtained by reacting ligand 1 with [Ru(η6-C6H6)Cl2]2 in the presence of NH4X salts, whereas neutral complex 4, Ru(η6-C6H6)(Cl)(fluorene-N[double bond, length as m-dash]CH-naphtholate), was isolated by reacting ligand 2 with the same precursor. It was possible to obtain a cationic version of the latter, 5[BF4], by reacting 4 with AgBF4 in the presence of pyridine. All compounds were fully characterized by NMR and HR-ESI-MS whereas some of them were also analyzed by single crystal X-ray analysis. Their in vitro antiproliferative activity was also assessed in human breast cancer cell lines, notably MCF-7 and T47D. Complex 4 and its cationic counterpart 5[BF4] were found to be the most cytotoxic compounds of the series (IC50 = 6.2-16.2 μM) and displayed higher antiproliferative activities than cisplatin in both cell lines. It was found that 5[BF4] undergoes a ligand exchange reaction and readily converts to 4 in the presence of 0.1 M NaCl, explaining the similarity in their observed cytotoxicities. Whereas 3[BF4] and 3[PF6] were found inactive at the tested concentrations, 3[BPh4] displayed a considerable cytotoxicity (IC50 = 16.7-27.8 μM). Notably, 3[BPh4], 4 (and 5[BF4]) were active against T47D, a cisplatin resistant cell line. Interestingly, 4 (16.4 μM) was found to be less cytotoxic than 3[BPh4] and cisplatin (6.6 and 7.9 μM, respectively) in breast healthy cells (MCF-12A). However, in comparison to 4 and cisplatin (at 10 μM), a lower in vivo toxicity was observed for complex 3[BPh4] on the development of zebrafish (Danio rerio) embryos.
Collapse
Affiliation(s)
- Mohammad Mehdi Haghdoost
- INRS - Centre Armand-Frappier Santé Biotechnology, Université du Québec, 531 boul. des Prairies, Laval, Quebec H7V 1B7, Canada.
| | - Golara Golbaghi
- INRS - Centre Armand-Frappier Santé Biotechnology, Université du Québec, 531 boul. des Prairies, Laval, Quebec H7V 1B7, Canada.
| | - Juliette Guard
- INRS - Centre Armand-Frappier Santé Biotechnology, Université du Québec, 531 boul. des Prairies, Laval, Quebec H7V 1B7, Canada.
| | - Sarah Sielanczyk
- INRS - Centre Armand-Frappier Santé Biotechnology, Université du Québec, 531 boul. des Prairies, Laval, Quebec H7V 1B7, Canada.
| | - Shunmoogum A Patten
- INRS - Centre Armand-Frappier Santé Biotechnology, Université du Québec, 531 boul. des Prairies, Laval, Quebec H7V 1B7, Canada.
| | - Annie Castonguay
- INRS - Centre Armand-Frappier Santé Biotechnology, Université du Québec, 531 boul. des Prairies, Laval, Quebec H7V 1B7, Canada.
| |
Collapse
|
37
|
Bomfim LM, de Araujo FA, Dias RB, Sales CBS, Rocha CAG, Correa RS, Soares MBP, Batista AA, Bezerra DP. Ruthenium(II) complexes with 6-methyl-2-thiouracil selectively reduce cell proliferation, cause DNA double-strand break and trigger caspase-mediated apoptosis through JNK/p38 pathways in human acute promyelocytic leukemia cells. Sci Rep 2019; 9:11483. [PMID: 31391500 PMCID: PMC6686011 DOI: 10.1038/s41598-019-47914-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/26/2019] [Indexed: 12/19/2022] Open
Abstract
Ruthenium(II) complexes with 6-methyl-2-thiouracil cis-[Ru(6m2tu)2(PPh3)2] (1) and [Ru(6m2tu)2(dppb)] (2) (where PPh3 = triphenylphosphine; dppb = 1,4-bis(diphenylphosphino)butane; and 6m2tu = 6-methyl-2-thiouracil) are potent cytotoxic agents and able to bind DNA. The aim of this study was to evaluate in vitro cellular underlying mechanism and in vivo effectiveness of these ruthenium(II) complexes in human acute promyelocytic leukemia HL-60 cells. Both complexes displayed potent and selective cytotoxicity in myeloid leukemia cell lines, and were detected into HL-60 cells. Reduction of the cell proliferation and augmented phosphatidylserine externalization, caspase-3, -8 and -9 activation and loss of mitochondrial transmembrane potential were observed in HL-60 cells treated with both complexes. Cotreatment with Z-VAD(OMe)-FMK, a pan-caspase inhibitor, reduced Ru(II) complexes-induced apoptosis. In addition, both metal complexes induced phosphorylation of histone H2AX (S139), JNK2 (T183/Y185) and p38α (T180/Y182), and cotreatment with JNK/SAPK and p38 MAPK inhibitors reduced complexes-induced apoptosis, indicating DNA double-strand break and activation of caspase-mediated apoptosis through JNK/p38 pathways. Complex 1 also reduced HL-60 cell growth in xenograft model. Overall, the outcome indicated the ruthenium(II) complexes with 6-methyl-2-thiouracil as a novel promising antileukemic drug candidates.
Collapse
Affiliation(s)
- Larissa M Bomfim
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | - Fênix A de Araujo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | - Caroline B S Sales
- Department of Biomorphology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-902, Brazil
| | | | - Rodrigo S Correa
- Department of Chemistry, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil.
| |
Collapse
|
38
|
Silva SLR, Baliza IRS, Dias RB, Sales CBS, Rocha CAG, Soares MBP, Correa RS, Batista AA, Bezerra DP. Ru(II)-thymine complex causes DNA damage and apoptotic cell death in human colon carcinoma HCT116 cells mediated by JNK/p38/ERK1/2 via a p53-independent signaling. Sci Rep 2019; 9:11094. [PMID: 31366902 PMCID: PMC6668648 DOI: 10.1038/s41598-019-47539-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/19/2019] [Indexed: 12/19/2022] Open
Abstract
Ru(II)-thymine complex [Ru(PPh3)2(Thy)(bipy)]PF6 (where PPh3 = triphenylphosphine, Thy = thyminate and bipy = 2,2′-bipyridine) is a potent cytotoxic agent with ability to bind to DNA, inducing caspase-mediated apoptosis in leukemia cells. In this study, we investigated the mechanism underlying the cell death induction by Ru(II)-thymine complex in human colon carcinoma HCT116 cells, as well as its effect in xenograft tumor model. The Ru(II)-thymine complex increased significantly the percentage of apoptotic HCT116 cells. Co-treatment with a JNK/SAPK inhibitor, p38 MAPK inhibitor and MEK inhibitor, which inhibit the activation of ERK1/2, caused a marked reduction of the percentage of complex-induced apoptotic cells. Moreover, the Ru(II)-thymine complex induced an increase in phospho-JNK2 (T183/Y185), phospho-p38α (T180/Y182) and phospho-ERK1 (T202/Y204) levels in HCT116 cells. Treatment with the Ru(II)-thymine complex increased significantly the phospho-histone H2AX (S139) expression, a DNA damage marker. The expression of phospho-p53 (S15) and MDM2 were not changed, and the co-treatment with a p53 inhibitor (cyclic pifithrin-α) did not reduce the complex-induced apoptosis in HCT116 cells, indicating that the Ru(II)-thymine complex induces DNA damage-mediated apoptosis by JNK/p38/ERK1/2 via a p53-independent signaling. The Ru(II)-thymine complex (1 and 2 mg/kg/day) also inhibited HCT116 cell growth in a xenograft model, reducing the tumor mass at 32.6–40.1%. Altogether, indicate that the Ru(II)-thymine complex is a promising anti-colon cancer drug candidate.
Collapse
Affiliation(s)
- Suellen L R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Ingrid R S Baliza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Caroline B S Sales
- Department of Biomorphology, Institute of Health Sciences, Federal University of Bahia, Salvador, Bahia, 40110-902, Brazil
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rodrigo S Correa
- Department of Chemistry, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
39
|
Sun JM, Xu HT, Zhao L, Zhang YB, Kang PC, Song ZF, Liu HS, Cui YF. Induction of cell-cycle arrest and apoptosis in human cholangiocarcinoma cells by pristimerin. J Cell Biochem 2019; 120:12002-12009. [PMID: 30825242 DOI: 10.1002/jcb.28485] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 01/24/2023]
Abstract
Pristimerin, a triterpenoid isolated from Celastraceae and Hippocrateaceae, is known to induce cytotoxicity in several cancer cell lines. However, whether pristimerin can induce apoptosis in cholangiocarcinoma cells and the underlying mechanism remain unexplored. We assessed the function of human cholangiocarcinoma QBC and RBE cell lines using various experimental methods such as the cell viability assay to elucidate the viability of cells, flow cytometry to detect the death rate of cells, and Western blot analysis to evaluate the expression of cell cycle-related proteins and autophagy-related proteins. Human cholangiocarcinoma QBC cells were transplanted to nude mice to establish an animal model, and the effect of pristimerin on tumor growth in this model was observed. QBC and RBE cell lines treated with pristimerin (0, 5, 10, and 20 μmol/L) demonstrated the induction of apoptosis in a dose-dependent manner. The cell viability assay revealed a reduction in the cell viability with an increase in the pristimerin concentration. Similarly, flow cytometry revealed a gradual increase in the cell death rate with an increase in the pristimerin concentration. In addition, pristimerin significantly lowered the expression of apoptosis-related proteins (Bcl-2, Bcl-xL, and procaspase-3), but increased the Bax expression. Furthermore, pristimerin resulted in the G0/G1 cell-cycle arrest, reducing the expression of cell cycle-related proteins (cyclin E, CDK2, and CDK4), and increased the expression of autophagy-related proteins (LC3) in QBC cell line. Treatment with pristimerin could inhibit tumor growth in the nude mouse model. Overall, this study suggests the potential effect of pristimerin on the cell-cycle arrest and apoptosis in human cholangiocarcinoma cells.
Collapse
Affiliation(s)
- Jian-Min Sun
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hai-Tao Xu
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Liang Zhao
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yu-Bao Zhang
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Peng-Cheng Kang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zeng-Fu Song
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hai-Shi Liu
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yun-Fu Cui
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
40
|
Liu Q, Xiao Y, Cai P, Li J, Li D. Long noncoding RNA DINO (damage induced noncoding) represses the development of gastric cancer by modulating p21 and Bcl-2 Associated X Protein (Bax) expression. J Cell Biochem 2019; 120:11190-11195. [PMID: 30775800 DOI: 10.1002/jcb.28394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
lncRNAs are responsible for a variety of diseases, including gastric cancer (GC). Many recent studies have reported that lncRNAs can serve as crucial regulators of various genes. Nevertheless, the biological function of lncRNA damage induced noncoding (DINO) remained poorly investigated in GC. Therefore, in our present study, the detailed role of DINO was investigated. It was manifested that DINO was significantly downregulated in GC tissues. Then, DINO was modulated by infecting LV-DINO or by LV-shRNA in BGC-823 and MGC-803 cells. Moreover, it was displayed that GC cell proliferation was suppressed by DINO overexpression, whereas silencing DINO increased cell proliferation significantly. For another, it was indicated that DINO dramatically induced apoptotic ratios of BGC-823 and MGC-803 cells, whereas the decrease of DINO depressed GC cell apoptosis. Apart from these, GC cell cycle progression was greatly blocked by LV-DINO. Furthermore, Western blot results displayed that upregulation of DINO elevated p21 expression and Bax expression. Oppositely, inhibition of DINO greatly suppressed p21 and Bax protein expression level. Taken these, DINO might exert a tumor inhibitory role in the progression of GC through modulating p21 and Bax.
Collapse
Affiliation(s)
- Qinqiang Liu
- Department of General Surgery, Xuzhou Hospital of Traditional Chinese medicine, Xuzhou, Jiangsu, China
| | - Yan Xiao
- Anaesthesiology department, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| | - Peng Cai
- Department of General Surgery, Xuzhou Hospital of Traditional Chinese medicine, Xuzhou, Jiangsu, China
| | - Jun Li
- Department of General Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dawei Li
- Department of General Surgery, Xuzhou Hospital of Traditional Chinese medicine, Xuzhou, Jiangsu, China
| |
Collapse
|
41
|
Qasim Warraich M, Ghion A, Perdisatt L, O'Neill L, Casey A, O'Connor C. In vitro cytotoxicity, cellular uptake, reactive oxygen species and cell cycle arrest studies of novel ruthenium(II) polypyridyl complexes towards A549 lung cancer cell line. Drug Chem Toxicol 2019; 44:319-329. [PMID: 30991856 DOI: 10.1080/01480545.2019.1589492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Ruthenium(II) polypyridyl complexes have displayed some promising biological responses against a variety of cancers and have emerged as a potential candidate that can show significant antitumor activity. Three ruthenium(II) polypyridyl complexes were biologically evaluated in vitro against the A549 cancer cell line. The complexes were selected based on initial DNA intercalation studies and MTT viability screening and were selected based on the most promising candidates, the [Ru(bpy)2o-CPIP].2PF6 (complex 1), [Ru(phen)2o-CPIP].2PF6 (complex 2) and [Ru(biq)2o-CPIP].2PF6 (complex 3). Confocal cellular uptake studies confirmed the intracellular transport of complexes into A549. Cytoplasmic and the nucleic accumulation of the complex 1 and 2 was seen while no fluorescent microscopy was performed for complex 3 due to instrumental limitations. Cellular cytotoxicity was investigated with the aid of the Alamar blue assay. The complexes displayed concentration and time dependent inhibitory effects yielding IC50 values from 5.00 to 32.75 µM. Complex 1 exhibit highest cytotoxicity with IC50 value of 5.00 ± 1.24 µM. All of the complexes have shown a significant effect in the reduction of intracellular reactive oxygen species (ROS) levels. Finally, the complexes have shown a transient effect on the cell cycle by arresting it at G0/G1 phase except for complex 2 [Ru(phen)2o-CPIP].2PF6 which has shown the significant G0/G1 arrest.
Collapse
Affiliation(s)
- Muhammad Qasim Warraich
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland.,School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
| | - Alessandra Ghion
- School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
| | - Laura Perdisatt
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland
| | - Luke O'Neill
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland
| | - Alan Casey
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland.,School of Physics, Clinical and Optometric Science, Technological University Dublin, Dublin, Ireland
| | - Christine O'Connor
- School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
| |
Collapse
|
42
|
De Grandis RA, Santos PWDSD, Oliveira KMD, Machado ART, Aissa AF, Batista AA, Antunes LMG, Pavan FR. Novel lawsone-containing ruthenium(II) complexes: Synthesis, characterization and anticancer activity on 2D and 3D spheroid models of prostate cancer cells. Bioorg Chem 2019; 85:455-468. [DOI: 10.1016/j.bioorg.2019.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/29/2019] [Accepted: 02/03/2019] [Indexed: 12/19/2022]
|
43
|
You Y, Wang N, He L, Shi C, Zhang D, Liu Y, Luo L, Chen T. Designing dual-functionalized carbon nanotubes with high blood–brain-barrier permeability for precise orthotopic glioma therapy. Dalton Trans 2019; 48:1569-1573. [PMID: 30499579 DOI: 10.1039/c8dt03948h] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Herein we synthesize a cell penetrating peptide- and cancer-targeted molecule-functionalized multi-walled carbon nanotube for precise orthotopic glioma therapy.
Collapse
Affiliation(s)
- Yuanyuan You
- The First Affiliated Hospital
- and Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Ni Wang
- The First Affiliated Hospital
- and Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Lizhen He
- The First Affiliated Hospital
- and Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Changzheng Shi
- The First Affiliated Hospital
- and Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Dong Zhang
- The First Affiliated Hospital
- and Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Yiyong Liu
- The First Affiliated Hospital
- and Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Liangping Luo
- The First Affiliated Hospital
- and Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Tianfeng Chen
- The First Affiliated Hospital
- and Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| |
Collapse
|
44
|
Zhao Z, Zhang X, Li CE, Chen T. Designing luminescent ruthenium prodrug for precise cancer therapy and rapid clinical diagnosis. Biomaterials 2018; 192:579-589. [PMID: 30551086 DOI: 10.1016/j.biomaterials.2018.12.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/20/2022]
Abstract
The effective design of a targeted drug delivery system could improve the therapeutic efficacy of anticancer drugs by reducing their undesirable adsorption and toxic side effects. Here, an RGD-peptide functionalized and bioresponsive ruthenium prodrug (Ru-RGD) was designed for both cancer therapy and clinical diagnosis. This prodrug can be selectively delivered to cervical tumor sites to enhance theranostic efficacy. The benzimidazole-based ligand of the complex is susceptible to acidic conditions so, after reaching the tumor microenvironment, ligand substitution occurs and the therapeutic drug is released. The deep-red emissions produced by both one-photon and two-photon excitation increases the potential of Ru-RGD for use in the deep tissue imaging of 3D tumor spheroids. The specific accumulation of the Ru prodrug in tumor sites allows for precise tumor diagnosis and therapy in vivo. Luminescence staining of 38 clinical patient specimens shows that Ru-RGD exhibits differences in binding capability between cervical cancer and normal tissue, with a sensitivity of 95% and a specificity of 100%. This study thus provides an approach for the effective design and application of targeted metal complexes in cancer therapy and clinical diagnosis.
Collapse
Affiliation(s)
- Zhennan Zhao
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Xiang Zhang
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Chang-E Li
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Chemistry, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
45
|
Lin K, Zhao ZZ, Bo HB, Hao XJ, Wang JQ. Applications of Ruthenium Complex in Tumor Diagnosis and Therapy. Front Pharmacol 2018; 9:1323. [PMID: 30510511 PMCID: PMC6252376 DOI: 10.3389/fphar.2018.01323] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/29/2018] [Indexed: 12/27/2022] Open
Abstract
Ruthenium complexes are a new generation of metal antitumor drugs that are currently of great interest in multidisciplinary research. In this review article, we introduce the applications of ruthenium complexes in the diagnosis and therapy of tumors. We focus on the actions of ruthenium complexes on DNA, mitochondria, and endoplasmic reticulum of cells, as well as signaling pathways that induce tumor cell apoptosis, autophagy, and inhibition of angiogenesis. Furthermore, we highlight the use of ruthenium complexes as specific tumor cell probes to dynamically monitor the active biological component of the microenvironment and as excellent photosensitizer, catalyst, and bioimaging agents for phototherapies that significantly enhance the diagnosis and therapeutic effect on tumors. Finally, the combinational use of ruthenium complexes with existing clinical antitumor drugs to synergistically treat tumors is discussed.
Collapse
Affiliation(s)
- Ke Lin
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zi-Zhuo Zhao
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hua-Ben Bo
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiao-Juan Hao
- Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | - Jin-Quan Wang
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
46
|
Aneesrahman KN, Rohini G, Bhuvanesh NSP, Sundararaj S, Musthafa M, Sreekanth A. In Vitro Biomolecular Interaction Studies and Cytotoxic Activities of Newly Synthesised Copper(II) Complexes Bearing 2-Hydroxynaphthaldehyde-Based Thiosemicarbazone. ChemistrySelect 2018. [DOI: 10.1002/slct.201800791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- K. N. Aneesrahman
- Department of Chemistry; National Institute of Technology; Tiruchirappalli 620015 India
| | - Gandhaveeti Rohini
- Department of Chemistry; National Institute of Technology; Tiruchirappalli 620015 India
| | | | | | - Moideen Musthafa
- Department of Chemistry; National Institute of Technology; Tiruchirappalli 620015 India
| | - Anandaram Sreekanth
- Department of Chemistry; National Institute of Technology; Tiruchirappalli 620015 India
| |
Collapse
|
47
|
Ru(II)-Thymine Complex Causes Cell Growth Inhibition and Induction of Caspase-Mediated Apoptosis in Human Promyelocytic Leukemia HL-60 Cells. Int J Mol Sci 2018; 19:ijms19061609. [PMID: 29848969 PMCID: PMC6032384 DOI: 10.3390/ijms19061609] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 04/15/2018] [Accepted: 05/02/2018] [Indexed: 01/01/2023] Open
Abstract
Ruthenium-based compounds represent a class of potential antineoplastic drugs. Recently, we designed, synthesized, and identified the Ru(II)-thymine complex [Ru(PPh3)2(Thy)(bipy)]PF6 (where PPh = triphenylphosphine, Thy = thymine and bipy = 2,2′-bipyridine) as a potent cytotoxic agent with the ability to bind to DNA and human and bovine serum albumins. In this study, the underlying cytotoxic mechanism of the [Ru(PPh3)2(Thy)(bipy)]PF6 complex was assessed. This complex displayed potent cytotoxicity in different cancer cell lines; the morphology that is associated with apoptotic cell death, increased internucleosomal DNA fragmentation without cell membrane permeability, loss of the mitochondrial transmembrane potential, increased phosphatidylserine externalization, and caspase-3 activation were observed in human promyelocytic leukemia HL-60 cells that were treated with the complex. Moreover, pretreatment of HL-60 cells with Z-VAD(OMe)-FMK, a pan-caspase inhibitor, partially reduced the apoptosis that was induced by the complex, indicating that the apoptotic cell death occurred through a caspase-mediated pathway. In conclusion, the [Ru(PPh3)2(Thy)(bipy)]PF6 complex displays potent cytotoxicity to different cancer cells and induces caspase-mediated apoptosis in HL-60 cells.
Collapse
|
48
|
Zeng D, Deng S, Sang C, Zhao J, Chen T. Rational Design of Cancer-Targeted Selenadiazole Derivative as Efficient Radiosensitizer for Precise Cancer Therapy. Bioconjug Chem 2018; 29:2039-2049. [PMID: 29771500 DOI: 10.1021/acs.bioconjchem.8b00247] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Delong Zeng
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Shulin Deng
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Chengcheng Sang
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Jianfu Zhao
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China
| |
Collapse
|
49
|
Photocytotoxic Copper(II) Complexes with Schiff-Base Scaffolds for Photodynamic Therapy. Chemistry 2018; 24:4111-4120. [DOI: 10.1002/chem.201705640] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Indexed: 01/27/2023]
|
50
|
Enhanced Tumor Diagnostic and Therapeutic Effect of Mesoporous Silica Nanoparticle-Mediated Pre-targeted Strategy. Pharm Res 2018; 35:63. [DOI: 10.1007/s11095-017-2338-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/20/2017] [Indexed: 12/29/2022]
|