1
|
Rogalewicz B, Czylkowska A. Recent advances in the discovery of copper(II) complexes as potential anticancer drugs. Eur J Med Chem 2025; 292:117702. [PMID: 40328033 DOI: 10.1016/j.ejmech.2025.117702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/13/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025]
Abstract
This review article offers a literature search of the most active, new copper (II) anticancer complexes based on nitrogen-containing ligands, reported in the literature over the past 5 years: from the beginning of 2019, until mid-2024. In the modern world, cancer remains one of the deadliest diseases of all. Although years of the ongoing research allowed us to better understand its nature, and thus aim more precisely at specific molecular targets and pathways, many of its aspects remain unclear. Today, chemotherapy still remains at the forefront of cancer treatment. With the ever-growing struggles to overcome chemoresistance and occurrence of serious side effects, the discovery of new, more selective and active drugs is a task of an utmost importance. At the same time, copper (II)-based compounds offer a wide array of biological activities and valuable biochemical properties. This review article provides the update on the recent advances in the discovery of new potential anticancer drugs among copper (II)-based compounds in the recent five years.
Collapse
Affiliation(s)
- Bartłomiej Rogalewicz
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924, Lodz, Poland.
| | - Agnieszka Czylkowska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924, Lodz, Poland.
| |
Collapse
|
2
|
Kazemi Z, Moini N, Rudbari HA, Micale N. A comprehensive review on the development of chiral Cu, Ni, and Zn complexes as pharmaceutical agents over the past decades: Synthesis, molecular structure and biological activity. Med Res Rev 2025; 45:654-754. [PMID: 39297288 DOI: 10.1002/med.22083] [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: 08/26/2022] [Revised: 07/09/2024] [Accepted: 08/25/2024] [Indexed: 02/06/2025]
Abstract
Chirality is a fundamental and widespread geometric structural property in living organisms that most biomacromolecules including nucleic acids, proteins and enzymes, possess. Consequently, the development of chiral drugs capable of binding specific targets have gradually gained wide attention in recent decades due to their selective effects on a broad spectrum of biological events ranging from cell metabolism to cell fate. In this context, the synthesis of chiral compounds as promising therapeutic candidates has assumed a major role in drug discovery. Among them, chiral metal complexes have attracted considerable interest due to their unique and intriguing structural features that could enable overcoming side effects and drug-resistance phenomena of metal-based drugs currently in the market such as cisplatin. In the current scenario, an in-depth overview of non-platinum chiral complexes needs to be presented and carried forward. Therefore, in this perspective article, an update of the scientific development of bioactive chiral copper, zinc and nickel complexes have been reported since they have not been thoroughly reviewed so far. Specifically, we focused the article mainly on metal complexes containing chiral ligands (type 2 chirality) as in literature they are more numerous than those with chirality at the metal center (type 1 chirality). Herein, not only their biological activity but also their mechanism of action is summarized. Furthermore, in the final section of the article we have highlighted copper-based complexes as those with a superior biological activity profile and greater prospects for development as a drug.
Collapse
Affiliation(s)
- Zahra Kazemi
- Department of Chemistry, University of Isfahan, Isfahan, Iran
| | - Nakisa Moini
- Department of Inorganic Chemistry, Faculty of Chemistry, Alzahra University, Tehran, Iran
| | | | - Nicola Micale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
3
|
Chowdhury M, Biswas N, Saha S, Rahaman A, Gupta PS, Banerjee A, Mandal DP, Bhattacharjee S, Zangrando E, Sciortino G, Pisanu F, Garribba E, Roy Choudhury R, Roy Choudhury C. Interaction with CT-DNA and in vitro cytotoxicity of two new copper(II)-based potential drugs derived from octanoic hydrazide ligands. J Inorg Biochem 2024; 256:112546. [PMID: 38593611 DOI: 10.1016/j.jinorgbio.2024.112546] [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: 09/25/2023] [Revised: 03/13/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024]
Abstract
Two copper(II) complexes [Cu(Hpmoh)(NO3)(NCS)] (1) and [Cu(peoh)(N3)]2 (2) were designed and synthesized by reaction of Cu(NO3)2·3H2O with hydrazone Schiff base ligands,abbreviated with Hpmoh and Hpeoh. Hpmoh and Hpeoh were prepared by condensation reaction of octanoic hydrazide with pyridine-2-carboxyaldehyde and 2-acetylpyridine, respectively. Complexes 1 and 2 were characterized using different analytical techniques such as FT-IR, UV-Vis, IR, EPR and single X-ray diffraction (XRD) analyses as well as computational methods (DFT). The XRD of 1 and 2 shows a mononuclear or a dinuclear structure with the copper(II) centre adopting a slightly distorted square pyramidal geometry. In water-containing solution and in DMSO, 1 and 2 undergo a partial transformation with formation of [Cu(Hpmoh)(NO3)(NCS)] (1) and [Cu(Hpmoh)(NO3)(H2O/DMSO)] (1a) in one system and [Cu(peoh)(N3)] (2a) in the other one, as supported by DFT calculations. Docking simulations confirmed that the intercalation is the preferred binding mode with DNA for 1, 1a and 2a, but suggested that the minor groove binding is also possible. A significant fluorescence quenching of the DNA-ethidium bromide conjugate was observed upon the addition of complexes 1 and 2 with a quenching constant around 104 M-1 s-1. Finally, both 1 and 2 were examined for anti-cancer activity using MDA-MB-231 (human breast adenocarcinoma) and A375 (malignant melanoma) cell lines through in vitro MTT assay which suggest comparable cancer cell killing efficacy, with the higher effectiveness of 2 due to the dissociation into two [Cu(peoh)(N3)] units.
Collapse
Affiliation(s)
- Manas Chowdhury
- Department of Chemistry, West Bengal State University, Barasat, Kolkata 700126, India
| | - Niladri Biswas
- Department of Biotechnology, Institute of Genetic Engineering, No. 30, Thakurhat Road, Badu, Madhyamgram, Kolkata, West Bengal 700128, India
| | - Sandeepta Saha
- Sripur High School, Madhyamgram Bazar, Kolkata 700130, India
| | - Ashikur Rahaman
- Department of Zoology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Poulami Sen Gupta
- Department of Zoology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Ankur Banerjee
- Department of Zoology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Deba Prasad Mandal
- Department of Zoology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Shamee Bhattacharjee
- Department of Zoology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Ennio Zangrando
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy
| | - Giuseppe Sciortino
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Federico Pisanu
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Eugenio Garribba
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, 07100 Sassari, Italy.
| | - Ruma Roy Choudhury
- Department of Chemistry and Environment, Heritage Institute of Technology, Chowbaga Road, Badu, Kolkata 700 107, India
| | | |
Collapse
|
4
|
Huang XQ, Wu RC, Liang JM, Zhou Z, Qin QP, Liang H. Anticancer activity of 8-hydroxyquinoline-triphenylphosphine rhodium(III) complexes targeting mitophagy pathways. Eur J Med Chem 2024; 272:116478. [PMID: 38718624 DOI: 10.1016/j.ejmech.2024.116478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/27/2024]
Abstract
Metallodrugs exhibiting distinct mechanisms of action compared with cisplatin hold promise for overcoming cisplatin resistance and improving the efficacy of anticancer drugs. In this study, a new series of rhodium (Rh)(III) complexes containing tris(triphenylphosphine)rhodium(I) chloride [(TPP)3RhCl] (TPP = triphenylphosphine, TPP=O = triphenylphosphine oxide) and 8-hydroxyquinoline derivatives (H-XR1-H-XR4), namely [Rh(XR1)2(TPP)Cl]·(TPP=O) (Yulin Normal University-1a [YNU-1a]), [Rh(XR2)2(TPP)Cl] (YNU-1b), [Rh(XR3)2(TPP)Cl] (YNU-1c), and [Rh(XR4)2(TPP)Cl] (YNU-1d), was synthesized and characterized via X-ray diffraction, mass spectrometry and IR. The cytotoxicity of the compounds YNU-1a-YNU-1d in Hep-G2 and HCC1806 human cancer cell lines and normal HL-7702 cell line was evaluated. YNU-1c exhibited cytotoxicity and selectivity in HCC1806 cells (IC50 = 0.13 ± 0.06 μM, selectivity factor (SF) = 384.6). The compounds YNU-1b and YNU-1c, which were selected for mechanistic studies, induced the activation of apoptotic pathways and mitophagy. In addition, these compounds released cytochrome c, cleaved caspase-3/pro-caspase-3 and downregulated the levels of mitochondrial respiratory chain complexes I/IV (M1 and M4) and ATP. The compound YNU-1c, which was selected for in vivo experiments, exhibited tumor growth inhibition (58.9 %). Importantly, hematoxylin and eosin staining and TUNEL revealed that HCC1806 tumor tissues exhibited significant apoptotic characteristics. YNU-1a-YNU-1d compounds are promising drug candidates that can be used to overcome cisplatin resistance.
Collapse
Affiliation(s)
- Xiao-Qiong Huang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Run-Chun Wu
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Jian-Min Liang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Zhen Zhou
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, PR China.
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, PR China.
| |
Collapse
|
5
|
Lu Y, Fan X, Pan Q, He B, Pu Y. A mitochondria-targeted anticancer copper dithiocarbamate amplifies immunogenic cuproptosis and macrophage polarization. J Mater Chem B 2024; 12:2006-2014. [PMID: 38291990 DOI: 10.1039/d3tb02886k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The way that cancer cells die inspires treatment regimens and cytolytic cuproptosis induced by copper complexes, like copper(II) bis(diethyldithiocarbamate) (CuET), has emerged as a novel therapeutic target. Herein, a triphenylphosphonium-modified CuET (TPP-CuET) is designed to target mitochondrial metabolism, triggering intense immunogenic cuproptosis in breast cancer cells and remodeling tumor-associated macrophages. TPP-CuET enables an enhanced mitochondrial copper accumulation in comparison to CuET (29.0% vs. 19.4%), and severely disrupts the morphology and functions of mitochondria, encompassing the tricarboxylic acid cycle, ATP synthesis, and electron transfer chain. Importantly, it triggers amplified immunogenic death of cancer cells, and the released damage-associated molecular patterns effectively induce M1 polarization and migration of macrophages. Transcriptome analysis further reveals that TPP-CuET promotes antigen processing and presentation in cancer cells through the MHC I pathway, activating the immune response of CD8 T cells and natural killer cells. To the best of our knowledge, TPP-CuET is the first mitochondrial targeted immunogenic cuproptosis inducer and is expected to flourish in antitumor immunotherapy.
Collapse
Affiliation(s)
- Yao Lu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China.
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
6
|
Du LQ, Zeng CJ, Mo DY, Qin QP, Tan MX, Liang H. 8-hydroxyquinoline-N-oxide copper(II)- and zinc(II)-phenanthroline and bipyridine coordination compounds: Design, synthesis, structures, and antitumor evaluation. J Inorg Biochem 2024; 251:112443. [PMID: 38100902 DOI: 10.1016/j.jinorgbio.2023.112443] [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: 09/29/2023] [Revised: 11/11/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023]
Abstract
Fourteen novel tumor-targeting copper(II) and zinc(II) complexes, [Cu(ONQ)(QD1)(NO3)]·CH3OH (NQ3), [Cu(ONQ)(QD2)(NO3)] (NQ2), [Cu(NQ)(QD2)Cl] (NQ3), [Cu(ONQ)(QD1)Cl] (NQ4), [Cu(ONQ)(QD3)](NO3) (NQ5), [Cu(ONQ)(QD3)Cl] (NQ6), [Zn(ONQ)(QD4)Cl] (NQ7), [Zn(ONQ)(QD1)Cl] (NQ8), [Zn(ONQ)(QD5)Cl] (NQ9), [Zn(ONQ)(QD2)Cl] (NQ10), [Zn(ONQ)(QD6)Cl] (NQ11), [Zn(ONQ)(QD7)Cl] (NQ12), and [Zn(ONQ)(QD3)Cl] (NQ13) supported on 8-hydroxyquinoline-N-oxide (H-ONQ), 2,2'-dipyridyl (QD1), 5,5'-dimethyl-2,2'-bipyridyl (QD2), 1,10-phenanthroline (QD3), 4,4'-dimethoxy-2,2'-bipyridyl (QD4), 4,4'-dimethyl-2,2'-bipyridyl (QD5), 5-chloro-1,10-phenanthroline (QD6), and bathophenanthroline (QD7), were first synthesized and characterized using various spectroscopic techniques. Furthermore, NQ1-NQ13 exhibited higher antiproliferative activity and selectivity for cisplatin-resistant SK-OV-3/DDP tumor cells (CiSK3) compared to normal HL-7702 cells based on results obtained from the cell counting Kit-8 (CCK-8) assay. The complexation of copper(II) ion with QD2 and ONQ ligands resulted in an evident increase in the antiproliferation of NQ1-NQ6, with NQ6 exhibiting the highest antitumor potency against CiSK3 cells compared to NQ1-NQ5, H-ONQ, QD1-QD7, and NQ7-NQ13 as well as the reference cisplatin drug with an IC50 value of 0.17 ± 0.05 μM. Mechanistic studies revealed that NQ4 and NQ6 induced apoptosis of CiSK3 cells via mitophagy pathway regulation and adenosine triphosphate (ATP) depletion. Further, the differential induction of mitophagy decreased in the order of NQ6 > NQ4, which can be attributed to the major impact of the QD3 ligand with a large planar geometry and the Cl leaving group within the NQ6 complex. In summary, these results confirmed that the newly synthesized H-ONQ copper(II) and zinc(II) coordination metal compounds NQ1-NQ13 exhibit potential as anticancer drugs for cisplatin-resistant ovarian CiSK3 cancer treatment.
Collapse
Affiliation(s)
- Ling-Qi Du
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Chu-Jie Zeng
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Dong-Yin Mo
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China.
| | - Ming-Xiong Tan
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China.
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China.
| |
Collapse
|
7
|
Zhang HQ, Lu X, Liang H, Chen ZF. Copper(II) complexes with plumbagin and bipyridines target mitochondria for enhanced chemodynamic cancer therapy. J Inorg Biochem 2024; 251:112432. [PMID: 38016329 DOI: 10.1016/j.jinorgbio.2023.112432] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 11/30/2023]
Abstract
The combination of mitochondrial targeting and chemodynamic therapy is a promising anti-cancer strategy. Three mitochondria targeting copper(II) complexes (Cu1-Cu3) with plumbagin and bipyridine ligands for enhanced chemodynamic therapy were synthesized and characterized. Their anti-proliferative activity to HeLa cells was higher than that of cisplatin, and their toxicity to normal cells was low. Cellular uptake and distribution studies indicated that Cu1 and Cu3 were mainly accumulated in mitochondria. The mechanism studies showed that Cu1 and Cu3 converted intracellular H2O2 into toxic hydroxyl radicals by consuming glutathione, leading to mitochondrial dysfunction. Treatment with the copper complex caused ER stress and cell arrest in the S phase which resulted in apoptosis. In vivo, Cu1 and Cu3 effectively inhibited the growth of HeLa xenograft tumors without obvious toxic and side effects.
Collapse
Affiliation(s)
- Hai-Qun 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xing Lu
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Zhen-Feng Chen
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
8
|
Xiong K, Lin X, Kou J, Wei F, Shen J, Chen Y, Ji L, Chao H. Apoferritin-Cu(II) Nanoparticles Induce Oncosis in Multidrug-Resistant Colon Cancer Cells. Adv Healthc Mater 2024; 13:e2302564. [PMID: 38073257 DOI: 10.1002/adhm.202302564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/03/2023] [Indexed: 12/19/2023]
Abstract
Multidrug resistance (MDR) limits the application of clinical chemotherapeutic drugs. There is an urgent need to develop non-apoptosis-inducing agents that circumvent drug resistance. Herein, four therapeutic copper complexes encapsulated in natural nanocarrier apoferritin (AFt-Cu1-4) are reported. Although they are isomers, they exhibit significantly different organelle distributions and cell death mechanisms. AFt-Cu1 and AFt-Cu3 accumulate in the cytoplasm and induce autophagy, whereas AFt-Cu2 and AFt-Cu4 can quickly enter the nucleus and trigger oncosis. Excitedly, AFt-Cu2 and AFt-Cu4 show a strong tumor growth inhibition effect in mice models bearing multidrug-resistant colon xenograft via intravenous injection. To the best of the authors' knowledge, this is the first example of metal-based nucleus-targeted oncosis inducers overcoming multidrug resistance in vivo.
Collapse
Affiliation(s)
- Kai Xiong
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Xinlin Lin
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Junfeng Kou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Fangmian Wei
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Jinchao Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
- MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 400201, P. R. China
| |
Collapse
|
9
|
Wang ZF, Huang XQ, Wu RC, Xiao Y, Zhang SH. Antitumor studies evaluation of triphenylphosphine ruthenium complexes with 5,7-dihalo-substituted-8-quinolinoline targeting mitophagy pathways. J Inorg Biochem 2023; 248:112361. [PMID: 37659141 DOI: 10.1016/j.jinorgbio.2023.112361] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/05/2023] [Accepted: 08/22/2023] [Indexed: 09/04/2023]
Abstract
Both ruthenium-containing complexes and 8-quinolinoline compounds have emerged as a potential novel agent for malignant tumor therapy. Here, three triphenylphosphine ruthenium complexes, [Ru(ZW1)(PPh3)2Cl2] (PPh3 = triphenylphosphine) (RuZ1), [Ru(ZW2)(PPh3)2Cl2] (RuZ2) and [Ru(ZW2)2(PPh3)Cl2]·CH2Cl2 (RuZ3) bearing 5,7-dichloro-8-quinolinol (H-ZW1) and 5,7-dichloro-8-hydroxyquinaldine (H-ZW2), have been synthesized, characterized and tested for their anticancer potential. We showed that triphenylphosphine ruthenium complexes RuZ1-RuZ3 impaired the cell viability of ovarian adenocarcinoma cisplatin-resistant SK-OV-3/DDP (SKO3CR) and SK-OV-3 (SKO3) cancer cells with greater selectivity and specificity than cisplatin. In addition, RuZ1-RuZ3 show higher excellent cytotoxicity than cisplatin towards SKO3CR cells, with IC50 values of 9.66 ± 1.08, 4.05 ± 0.67 and 7.18 ± 0.40 μM, respectively, in which the SKO3CR cells was the most sensitive to RuZ1-RuZ3. Depending on the substituent type, the antiproliferative ability of RuZ1-RuZ3 followed the trend: -CH3 > -H. However, RuZ1-RuZ3 have no obvious toxicity to normal cell HL-7702. Besides, RuZ1 and RuZ2 could induce mitophagy related-apoptosis pathways through suppression of mitochondrial membrane potential (ΔΨm), accumulation of [Ca2+] and reactive oxygen species (ROS), and regulation of LC3 II/LC3 I, Beclin-1, P62, FUNDC1, PINK1, Parkin, cleaved-caspase-3, caspase-9 and cytochrome c signaling pathway, and hindering the preparation of mitochondrial respiration complexes I and IV and ATP levels. Mechanistic study revealed that RuZ1 and RuZ2 induce apoptosis in SKO3CR cells via mitophagy related-apoptosis pathways induction and energy (ATP) generation disturbance. Taken together, the studied triphenylphosphine ruthenium complexes RuZ1-RuZ3 are promising chemotherapeutic agents with high effectiveness and low toxicity.
Collapse
Affiliation(s)
- Zhen-Feng Wang
- College of Chemistry, Guangdong University of Petrochemical Technology, Maoming, Guangdong, PR China; Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, Guilin University of Technology, Guilin, PR China
| | - Xiao-Qiong Huang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Run-Chun Wu
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Yu Xiao
- College of Chemistry, Guangdong University of Petrochemical Technology, Maoming, Guangdong, PR China.
| | - Shu-Hua Zhang
- College of Chemistry, Guangdong University of Petrochemical Technology, Maoming, Guangdong, PR China; Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, Guilin University of Technology, Guilin, PR China.
| |
Collapse
|
10
|
Ma FJ, Huang X, Li XY, Tang SL, Li DJ, Cheng YZ, Azam M, Zhang LP, Sun D. Synthesis, structure and biological activity of silver(I) complexes containing triphenylphosphine and non-steroidal anti-inflammatory drug ligands. J Inorg Biochem 2023; 250:112404. [PMID: 39492372 DOI: 10.1016/j.jinorgbio.2023.112404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/05/2024]
Abstract
Two Ag(I) complexes containing triphenylphosphine and non-steroidal anti-inflammatory drug ligands were synthesized and investigated using various spectroscopic studies and single crystal X-ray crystallography. The binding properties of tolfenamic acid, ibuprofen and the two complexes with DNA and BSA were investigated using UV or fluorescence spectroscopy. The results showed that two Ag(I) complexes bound to DNA by the intercalation mode and interacted with BSA using a static quenching procedure. Furthermore, the results of fluorescence titration suggested that the complexes had good affinity for BSA and one binding site close to BSA. The in vitro cytotoxicity of tolfenamic acid, ibuprofen, and the two complexes against four human carcinoma cell lines (MCF-7, HepG-2, A549, and HeLa cells) was tested using an MTT assay. Complex 1 had higher cytotoxicity against HeLa cells. The intracellular reactive oxygen species (ROS) assay showed complex 1 induced the ROS generation in HeLa cells in a concentration dependent manner. Flow cytometry analysis showed complex 1 could suppress the HeLa cells growth during the G0/G1 phase and induce apoptosis in dose-depended manner.
Collapse
Affiliation(s)
- Feng-Jie Ma
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Xiang Huang
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Xue-Ying Li
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Shi-Li Tang
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - De-Jun Li
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Yuan-Zheng Cheng
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China.
| | - Mohammad Azam
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Li-Ping Zhang
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China.
| | - Di Sun
- School of Chemistry and Chemical Engineering, State Key Laboratory of Crystal Materials, Shandong University, Ji'nan 250100, PR China.
| |
Collapse
|
11
|
Hata M, Ueno J, Hitomi Y, Kodera M. Roles of DNA Target in Cancer Cell-Selective Cytotoxicity by Dicopper Complexes with DNA Target/Ligand Conjugates. ACS OMEGA 2023; 8:28690-28701. [PMID: 37576680 PMCID: PMC10413468 DOI: 10.1021/acsomega.3c03387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023]
Abstract
The DNA target/ligand conjugates (HLX, X = Pn and Mn, n = 1-3) were synthesized where various lengths of -CONH(CH2CH2O)nCH2CH2NHCO- linkers with a 9-phenanthrenyl (P) or methyl (M) terminal as DNA targets replace the methyl group of 2,6-di(amide-tether cyclen)-p-cresol ligand (HL). DNA binding, DNA cleavage, cellular uptake, and cytotoxicity of [Cu2(μ-OH)(LX)](ClO4)2 (1X) are examined and compared with those of [Cu2(μ-OH)(L)](ClO4)2 (1) to clarify roles of DNA targets. Upon reaction of 1X with H2O2, μ-1,1-O2H complexes are formed for DNA cleavage. 1P1, 1P2, and 1P3 are 22-, 11-, 3-fold more active for conversion of Form II to III in the cleavage of supercoiled plasmid DNA with H2O2 than 1, where the short P-linker may fix a dicopper moiety within a small number of base pairs to facilitate DNA double-strand breaks (dsb). This enhances the proapoptotic activity of 1P1, 1P2, and 1P3, which are 30-, 12-, and 9.9-fold cytotoxic against HeLa cells than 1. DNA dsb and cytotoxicity are 44% correlated in 1P1-3 but 5% in 1M1-3, suggesting specific DNA binding of P-linkers and nonspecific binding of M-linkers in biological cells. 1P1-3 exert cancer cell-selective cytotoxicity against lung and pancreas cancer and normal cells where the short P-linker enhances the selectivity, but 1M1-3 do not. Intracellular visualization, apoptosis assay, and caspase activity assay clarify mitochondrial apoptosis caused by 1P1-3. The highest cancer cell selectivity of 1P1 may be enabled by the short P-linker promoting dsb of mitochondrial DNA with H2O2 increased by mitochondrial dysfunction in cancer cells.
Collapse
Affiliation(s)
- Machi Hata
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Jin Ueno
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Yutaka Hitomi
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Masahito Kodera
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| |
Collapse
|
12
|
Cai DH, Liang BF, Chen BH, Liu QY, Pan ZY, Le XY, He L. A novel water-soluble Cu(II) gluconate complex inhibits cancer cell growth by triggering apoptosis and ferroptosis related mechanisms. J Inorg Biochem 2023; 246:112299. [PMID: 37354603 DOI: 10.1016/j.jinorgbio.2023.112299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
Metal copper complexes have attracted extensive attention as potential alternatives to platinum-based anticancer drugs due to their possible different modes of action. Herein, a new copper(II) gluconate complex, namely [Cu(DPQ)(Gluc)]·2H2O (CuGluc, DPQ = pyrazino[2,3-f][1,10]phenanthroline), with good water-solubility and high anticancer activity was synthesized by using D-gluconic acid (Gluc-2H) as an auxiliary ligand. The complex was well characterized by single-crystal X-ray diffraction analysis, elemental analysis, molar conductivity, and Fourier transform infrared spectroscopy (FTIR). The DNA-binding experiments revealed that CuGluc was bound to DNA by intercalation with end-stacking binding. CuGluc could oxidatively cleave DNA, in which 1O2 and H2O2 were involved. In addition, CuGluc was bound to the IIA subdomain of human serum albumin (HSA) through hydrophobic interaction and hydrogen bonding, showing a good affinity for HSA. The complex showed superior anticancer activity toward several cancer cells than cisplatin in vitro. Further studies indicated that CuGluc caused apoptotic cell death in human liver cancer (HepG2) cells through elevated intracellular reactive oxygen species (ROS) levels, mitochondrial dysfunction, cell cycle arrest, and caspase activation. Interestingly, CuGluc also triggered the ferroptosis mechanism through lipid peroxide accumulation and inhibition of glutathione peroxidase 4 (GPX4) activity. More importantly, CuGluc significantly inhibited tumor growth in vivo, which may benefit from the combined effects of apoptosis and ferroptosis. This work provides a promising strategy to develop highly effective antitumor copper complexes by coordinating with the glucose metabolite D-gluconic acid and exploiting the synergistic effects of apoptosis and ferroptosis mechanisms.
Collapse
Affiliation(s)
- Dai-Hong Cai
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Bin-Fa Liang
- School of Pharmaceutical Sciences, Medical School, Shenzhen University, Shenzhen 518060, China
| | - Bai-Hua Chen
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Qi-Yan Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Zheng-Yin Pan
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| | - Xue-Yi Le
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| | - Liang He
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
13
|
Alfadul SM, Matnurov EM, Varakutin AE, Babak MV. Metal-Based Anticancer Complexes and p53: How Much Do We Know? Cancers (Basel) 2023; 15:2834. [PMID: 37345171 DOI: 10.3390/cancers15102834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
P53 plays a key role in protecting the human genome from DNA-related mutations; however, it is one of the most frequently mutated genes in cancer. The P53 family members p63 and p73 were also shown to play important roles in cancer development and progression. Currently, there are various organic molecules from different structural classes of compounds that could reactivate the function of wild-type p53, degrade or inhibit mutant p53, etc. It was shown that: (1) the function of the wild-type p53 protein was dependent on the presence of Zn atoms, and (2) Zn supplementation restored the altered conformation of the mutant p53 protein. This prompted us to question whether the dependence of p53 on Zn and other metals might be used as a cancer vulnerability. This review article focuses on the role of different metals in the structure and function of p53, as well as discusses the effects of metal complexes based on Zn, Cu, Fe, Ru, Au, Ag, Pd, Pt, Ir, V, Mo, Bi and Sn on the p53 protein and p53-associated signaling.
Collapse
Affiliation(s)
- Samah Mutasim Alfadul
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Egor M Matnurov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Alexander E Varakutin
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| |
Collapse
|
14
|
Yang Y, Guo FF, Chen CF, Li YL, Liang H, Chen ZF. Antitumor activity of synthetic three copper(II) complexes with terpyridine ligands. J Inorg Biochem 2023; 240:112093. [PMID: 36525715 DOI: 10.1016/j.jinorgbio.2022.112093] [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: 09/24/2022] [Revised: 12/03/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Three new synthetic terpyridine copper(II) complexes were characterized. The copper(II) complexes induced apoptosis of three cancer cell lines and arrested T-24 cell cycle in G1 phase. The complexes were accumulated in mitochondria of T-24 cells and caused significant reduction of the mitochondrial membrane potential. The complexes increased both intracellular ROS and Ca2+ levels and activated the caspase-3/9 expression. The apoptosis was further confirmed by Western Blotting analysis. Bcl-2 was down-regulated and Bax was upregulated after treatment with complexes 1-3. The in vivo studies showed that complexes 1-3 obviously inhibited the growth of tumor without significant toxicity to other organs.
Collapse
Affiliation(s)
- Yang 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; Department of Chemistry and Pharmacy, Guilin Normal College, Guilin 541199, China
| | - Fei-Fei Guo
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Cai-Feng Chen
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yu-Lan 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Zhen-Feng Chen
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
15
|
Chen X, Jia Z, Wen Y, Huang Y, Yuan X, Chen Y, Liu Y, Liu J. Bidirectional anisotropic palladium nanozymes reprogram macrophages to enhance collaborative chemodynamic therapy of colorectal cancer. Acta Biomater 2022; 151:537-548. [PMID: 35981687 DOI: 10.1016/j.actbio.2022.08.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/17/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
Abstract
In the complex tumor microenvironment (TME), tumor-associated macrophages (TAMs) play an important role in immunosuppression and tumor growth; hence, tumor cells are no longer the only target during tumor treatment. However, how to simultaneously target both tumor cells and TAMs to effectively eliminate the tumor remains a challenge. Herein, based on the specific receptors for cancer cells and TAMs, we prepared bidirectional anisotropic palladium nanoclusters (Pd-HA+Pd-M@R NPs) to simultaneously target tumor cells and TAMs for enhancing the therapeutic effect. In these nanoclusters, the Pd-HA part was obtained by modifying hyaluronic acid (HA) on the surface of ultra-small Pd nanozymes that could target CT26 cells. Moreover, with the high peroxidase (POD) and catalase (CAT) activity of Pd nanozymes, Pd-HA NPs directly caused cancer cell death by producing H2O2 and highly toxic reactive oxygen therapy (ROS) through chemodynamic therapy (CDT). The other part of Pd NPs functioned as a carrier that linked mannose (Man) and the imiquimod molecule (R837) to obtain Pd-M@R NPs, which could specifically connect the mannose receptor of TAMs and perform targeted reprogramming of TAMs to M1 phenotype to reverse immunosuppression and further activate immunotherapy to form "double therapy". Therefore, the strategy of "double therapy" provides new sights for treating malignant tumors. STATEMENT OF SIGNIFICANCE: The bidirectional anisotropic Pd nanoclusters (Pd-HA+Pd-M@R NPs) that can simultaneously target the tumor cells and TAMs with the modification of HA and mannose, respectively. Under the biodirectional anisotropic effect, the Pd nanozymes in Pd-HA can directly kill CT 26 cells through catalyze producing toxic ROS. The Pd-M@R exhibited effectively delivery the imiquimod molecule (R837) to TAMs and specifically induced it transformed into M1 phenotype to reverse tumor immunosuppression to form the "double therapy".
Collapse
Affiliation(s)
- Xu Chen
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Zhi Jia
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Yayu Wen
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Yuqin Huang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Xiaoyu Yuan
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Yutong Chen
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Yanan Liu
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China
| | - Jie Liu
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
16
|
Kuznetcova I, Bacher F, Alfadul SM, Tham MJR, Ang WH, Babak MV, Rapta P, Arion VB. Elucidation of Structure-Activity Relationships in Indolobenzazepine-Derived Ligands and Their Copper(II) Complexes: the Role of Key Structural Components and Insight into the Mechanism of Action. Inorg Chem 2022; 61:10167-10181. [PMID: 35713376 PMCID: PMC9490829 DOI: 10.1021/acs.inorgchem.2c01375] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Indolo[3,2-d][1]benzazepines (paullones), indolo[3,2-d][2]benzazepines, and indolo[2,3-d][2]benzazepines (latonduines) are isomeric scaffolds of current medicinal interest. Herein, we prepared a small library of novel indolo[3,2-d][2]benzazepine-derived ligands HL1-HL4 and copper(II) complexes 1-4. All compounds were characterized by spectroscopic methods (1H and 13C NMR, UV-vis, IR) and electrospray ionization (ESI) mass spectrometry, while complexes 2 and 3, in addition, by X-ray crystallography. Their purity was confirmed by HPLC coupled with high-resolution ESI mass spectrometry and/or elemental analysis. The stability of compounds in aqueous solutions in the presence of DMSO was confirmed by 1H NMR and UV-vis spectroscopy measurements. The compounds revealed high antiproliferative activity in vitro in the breast cancer cell line MDA-MB-231 and hepatocellular carcinoma cell line LM3 in the low micromolar to nanomolar concentration range. Important structure-activity relationships were deduced from the comparison of anticancer activities of HL1-HL4 and 1-4 with those of structurally similar paullone-derived (HL5-HL7 and 5-7) and latonduine-derived scaffolds (HL8-HL11 and 8-11). The high anticancer activity of the lead drug candidate 4 was linked to reactive oxygen species and endoplasmic reticulum stress induction, which were confirmed by fluorescent microscopy and Western blot analysis.
Collapse
Affiliation(s)
- Irina Kuznetcova
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Felix Bacher
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Samah Mutasim Alfadul
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Max Jing Rui Tham
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117544, Singapore
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117544, Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Peter Rapta
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, SK-81237 Bratislava, Slovak Republic
| | - Vladimir B Arion
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| |
Collapse
|
17
|
Bergamini FR, Nunes JH, Manzano CM, de Carvalho MA, Ribeiro MA, Ruiz ALTG, de Carvalho JE, Lustri WR, de Paiva REF, Portes MC, da Costa Ferreira AM, Corbi PP. Investigating the antiproliferative activities of new CuII complexes with pyridine hydrazone derivatives of nalidixic acid. J Inorg Biochem 2022; 234:111881. [DOI: 10.1016/j.jinorgbio.2022.111881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/10/2022] [Accepted: 05/28/2022] [Indexed: 11/16/2022]
|
18
|
Chen YT, Zhang SN, Wang ZF, Wei QM, Zhang SH. Discovery of thirteen cobalt(II) and copper(II) salicylaldehyde Schiff base complexes that induce apoptosis and autophagy in human lung adenocarcinoma A549/DDP cells and that can overcome cisplatin resistance in vitro and in vivo. Dalton Trans 2022; 51:4068-4078. [PMID: 35179159 DOI: 10.1039/d1dt03749h] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this study, 13 transition metal complexes, namely, [Cu(L1H)(H2O)2]·(H2O)·NO3 (1), [Cu(LnH2)2]·(NO3)·(H2O)2 (2, n = 2; 3, n = 3; 4, n = 4; 5, n = 5), [Co(LnH)2]2·(H2O)0.5 (6, n = 2; 7, n = 3; 8, n = 4; 9, n = 5), [Cu(L6H)0.5(L10H)0.5(phen)]·(CH3OH)0.25 (10), [Cu(L11H) (phen)]4·(H2O)9 (11), [Cu(L8H)0.27(L12H)0.73(phen)]4·(H2O)5.5(CH3OH) (12), and [Cu(L9H) (phen)]3·(H2O)7·(CH3OH) (13), were synthesized using Schiff base ligands and characterized by elemental analysis (EA), infrared spectroscopy (IR), and single-crystal X-ray diffraction (SC-XRD). Compared with complexes 1-9, complexes 10-13 displayed stronger cytotoxic activities against the tested A549/DDP cancer cells (IC50 = 0.97-3.31 μM), with differences greater than one order of magnitude. Moreover, complexes 11 and 13 could induce apoptosis and autophagy in A549/DDP cells via the mitochondrial dysfunction pathway that affects the regulation of autophagy- and mitochondrial-related proteins. Importantly, the results indicate that the two novel salicylaldehyde Schiff base analogs, 11 and 13, exhibited pronounced and selective activity against A549/DDP xenografts in vivo.
Collapse
Affiliation(s)
- Ya-Ting Chen
- College of Chemistry, Guangdong University of Petrochemical Technology, Maoming, Guangdong, 525000, P R China. .,Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, P R China
| | - Shao-Nan Zhang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, P R China
| | - Zhen-Feng Wang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, P R China
| | - Qing-Min Wei
- College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Shu-Hua Zhang
- College of Chemistry, Guangdong University of Petrochemical Technology, Maoming, Guangdong, 525000, P R China. .,Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, P R China
| |
Collapse
|
19
|
Gu YQ, Zhong YJ, Hu MQ, Li HQ, Yang K, Dong Q, Liang H, Chen ZF. Terpyridine copper(II) complexes as potential anticancer agents by inhibiting cell proliferation, blocking the cell cycle and inducing apoptosis in BEL-7402 cells. Dalton Trans 2022; 51:1968-1978. [PMID: 35023532 DOI: 10.1039/d1dt02988f] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Four mononuclear terpyridine complexes [Cu(H-La)Cl2]·CH3OH (1), [Cu(H-La)Cl]ClO4 (2), [Cu(H-Lb)Cl2]·CH3OH (3), and [Cu(H-Lb)(CH3OH)(DMSO)](ClO4)2 (4) were prepared and fully characterized. Complexes 1-4 exhibited higher cytotoxic activity against several tested cancer cell lines especially BEL-7402 cells compared to cisplatin, and they showed low toxicity towards normal human liver cells. ICP-MS detection indicated that the copper complexes were accumulated in mitochondria. Mechanistic studies demonstrated that the copper complexes induced G0/G1 arrest and altered the expression of the related proteins of the cell cycle. All copper complexes reduced the mitochondrial membrane potential while increasing the intracellular ROS levels and the release of Ca2+. They also up-regulated Bax and down-regulated Bcl-2 expression levels, caused cytochrome c release and the activation of the caspase cascade, and induced mitochondrion-mediated apoptosis. Animal studies demonstrated that complex 1 suppressed tumor growth in a mouse xenograft model bearing BEL-7402 tumor cells.
Collapse
Affiliation(s)
- Yun-Qiong Gu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Centre for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China. .,School of Environment and Life Science, Nanning Normal University, Nanning, 530001, P. R China
| | - Yu-Jun Zhong
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Centre for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China.
| | - Mei-Qi Hu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Centre for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China.
| | - Huan-Qing Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Centre for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China.
| | - Kun Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Centre for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China.
| | - Qi Dong
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Centre for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China.
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Centre for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China.
| | - Zhen-Feng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Centre for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China.
| |
Collapse
|
20
|
Cai DH, Chen BH, Liu QY, Le XY, He L. Synthesis, structural studies, interaction with DNA/HSA and antitumor evaluation of new Cu( ii) complexes containing 2-(1 H-imidazol-2-yl)pyridine and amino acids. Dalton Trans 2022; 51:16574-16586. [DOI: 10.1039/d2dt02985e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
New Cu(ii) complexes with promising anticancer activity induce apoptosis in HepG2 cells through DNA damage and cytotoxic ROS-mediated mitochondrial dysfunction pathways.
Collapse
Affiliation(s)
- Dai-Hong Cai
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
- Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| | - Bai-Hua Chen
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
- Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| | - Qi-Yan Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
- Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| | - Xue-Yi Le
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
- Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| | - Liang He
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
- Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
21
|
Jana K, Pramanik U, Ingle KS, Maity R, Mukherjee S, Nayak SK, Chandra Debnath S, Maity T, Maity S, Chandra Samanta B. Copper(II) complexes with NNN and NNO Schiff base ligands as efficient photodegradation agents for methylene blue, preferential BSA binder and biomaterial transplants. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2021.113565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
22
|
Ma H, Long S, Cao J, Xu F, Zhou P, Zeng G, Zhou X, Shi C, Sun W, Du J, Han K, Fan J, Peng X. New Cy5 photosensitizers for cancer phototherapy: a low singlet-triplet gap provides high quantum yield of singlet oxygen. Chem Sci 2021; 12:13809-13816. [PMID: 34760166 PMCID: PMC8549779 DOI: 10.1039/d1sc04570a] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/21/2021] [Indexed: 01/12/2023] Open
Abstract
Highly efficient triplet photosensitizers (PSs) have attracted increasing attention in cancer photodynamic therapy where photo-induced reactive oxygen species (ROSs, such as singlet oxygen) are produced via singlet–triplet intersystem crossing (ISC) of the excited photosensitizer to kill cancer cells. However, most PSs exhibit the fatal defect of a generally less-than-1% efficiency of ISC and low yield of ROSs, and this defect strongly impedes their clinical application. In the current work, a new strategy to enhance the ISC and high phototherapy efficiency has been developed, based on the molecular design of a thio-pentamethine cyanine dye (TCy5) as a photosensitizer. The introduction of an electron-withdrawing group at the meso-position of TCy5 could dramatically reduce the singlet–triplet energy gap (ΔEst) value (from 0.63 eV to as low as 0.14 eV), speed up the ISC process (τISC = 1.7 ps), prolong the lifetime of the triplet state (τT = 319 μs) and improve singlet oxygen (1O2) quantum yield to as high as 99%, a value much higher than those of most reported triplet PSs. Further in vitro and in vivo experiments have shown that TCy5-CHO, with its efficient 1O2 generation and good biocompatibility, causes an intense tumor ablation in mice. This provides a new strategy for designing ideal PSs for cancer photo-therapy. The electron-withdrawing group at the meso-position of Thio-Cy5 could dramatically reduce the singlet–triplet energy gap, and speed up the intersystem crossing process.![]()
Collapse
Affiliation(s)
- He Ma
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China
| | - Saran Long
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China .,State Key Laboratory of Fine Chemicals and Shenzhen Research Institute, Dalian University of Technology Dalian 116024 China
| | - Jianfang Cao
- School of Chemical Engineering, Dalian University of Technology Panjin Campus Panjin 124221 China
| | - Feng Xu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China
| | - Panwang Zhou
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University Qingdao 266237 China
| | - Guang Zeng
- State Key Laboratory of Catalysis, Dalian Institute of Chemical and Physics, Chinese Academy of Sciences Zhongshan Road 457 Dalian 116023 China
| | - Xiao Zhou
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China
| | - Chao Shi
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China .,State Key Laboratory of Fine Chemicals and Shenzhen Research Institute, Dalian University of Technology Dalian 116024 China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China .,State Key Laboratory of Fine Chemicals and Shenzhen Research Institute, Dalian University of Technology Dalian 116024 China
| | - Keli Han
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences 457, Zhongshan Road Dalian 116023 China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China .,State Key Laboratory of Fine Chemicals and Shenzhen Research Institute, Dalian University of Technology Dalian 116024 China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 China .,State Key Laboratory of Fine Chemicals and Shenzhen Research Institute, Dalian University of Technology Dalian 116024 China
| |
Collapse
|
23
|
Tsakanova G, Stepanyan A, Arakelova E, Ayvazyan V, Tonoyan V, Arakelyan A, Hildebrandt G, Schültke E. The radioenhancement potential of Schiff base derived copper (II) compounds against lung carcinoma in vitro. PLoS One 2021; 16:e0253553. [PMID: 34143847 PMCID: PMC8213134 DOI: 10.1371/journal.pone.0253553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/07/2021] [Indexed: 11/30/2022] Open
Abstract
For the last years, copper complexes have been intensively implicated in biomedical research as components of cancer treatment. Herewith, we provide highlights of the synthesis, physical measurements, structural characterization of the newly developed Cu(II) chelates of Schiff Bases, Cu(Picolinyl-L-Tryptopahanate)2, Cu(Picolinyl-L-Tyrosinate)2, Cu(Isonicotinyl-L-Tyrosinate)2, Cu(Picolinyl-L-Phenylalaninate)2, Cu(Nicotinyl-L-Phenylalaninate)2, Cu(Isonicotinyl-L-Phenylalaninate)2, and their radioenhancement capacity at kV and MV ranges of irradiation of human lung carcinoma epithelial cells in vitro. The methods of cell growth, viability and proliferation were used. All compounds exerted very potent radioenhancer capacities in the irradiated lung carcinoma cells at both kV and MV ranges in a 100 μM concentration. At a concentration of 10 μM, only Cu(Picolinyl-L-Tyrosinate)2, Cu(Isonicotinyl-L-Tyrosinate)2, Cu(Picolinyl-L-Phenylalaninate)2 possessed radioenhancer properties at kV and MV ranges. Cu(Picolinyl-L-Tryptophanate)2 showed radioenhancer properties only at kV range. Cu(Nicotinyl-L-Phenylalaninate)2 and Cu(Isonicotinyl-L-Phenylalaninate)2 showed remarkable radioenhancer activity only at MV range. All compounds acted in dose-dependent manner at both tested energy ranges. These copper (II) compounds, in combination with 1 Gy irradiation at either 120 kV or 6 MV, are more efficient at delaying cell growth of lung cancer cells and at reducing cell viability in vitro than the irradiation administered alone. Thus, we have demonstrated that the studied copper compounds have a good potential for radioenhancement.
Collapse
Affiliation(s)
- Gohar Tsakanova
- Institute of Molecular Biology NAS RA, Yerevan, Armenia
- CANDLE Synchrotron Research Institute, Yerevan, Armenia
| | - Ani Stepanyan
- Institute of Molecular Biology NAS RA, Yerevan, Armenia
| | | | | | - Vahan Tonoyan
- Institute of Molecular Biology NAS RA, Yerevan, Armenia
| | | | - Guido Hildebrandt
- Department of Radiooncology, Rostock University Medical Center, Rostock, Germany
| | - Elisabeth Schültke
- Department of Radiooncology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
24
|
Wang H, Fang B, Peng B, Wang L, Xue Y, Bai H, Lu S, Voelcker NH, Li L, Fu L, Huang W. Recent Advances in Chemical Biology of Mitochondria Targeting. Front Chem 2021; 9:683220. [PMID: 34012953 PMCID: PMC8126688 DOI: 10.3389/fchem.2021.683220] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are vital subcellular organelles that generate most cellular chemical energy, regulate cell metabolism and maintain cell function. Mitochondrial dysfunction is directly linked to numerous diseases including neurodegenerative disorders, diabetes, thyroid squamous disease, cancer and septicemia. Thus, the design of specific mitochondria-targeting molecules and the realization of real-time acquisition of mitochondrial activity are powerful tools in the study and treatment of mitochondria dysfunction in related diseases. Recent advances in mitochondria-targeting agents have led to several important mitochondria chemical probes that offer the opportunity for selective targeting molecules, novel biological applications and therapeutic strategies. This review details the structural and physiological functional characteristics of mitochondria, and comprehensively summarizes and classifies mitochondria-targeting agents. In addition, their pros and cons and their related chemical biological applications are discussed. Finally, the potential biomedical applications of these agents are briefly prospected.
Collapse
Affiliation(s)
- Haiwei Wang
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Bin Fang
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
- School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Limin Wang
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Yufei Xue
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Shenci Lu
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Nicolas H. Voelcker
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, Australia
- Department of Materials Science & Engineering, Monash University, Clayton, VIC, Australia
| | - Lin Li
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
| | - Li Fu
- School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics, Xi’an Institute of Flexible Electronics (IFE) and Xi’an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi’an, China
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, China
| |
Collapse
|
25
|
Xiong K, Zhou Y, Lin X, Kou J, Lin M, Guan R, Chen Y, Ji L, Chao H. Cyclometalated Iridium(III) Complexes as Mitochondria-targeting Photosensitizers against Cisplatin-resistant Cells †. Photochem Photobiol 2021; 98:85-91. [PMID: 33617666 DOI: 10.1111/php.13404] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/01/2022]
Abstract
Four iridium (III) complexes Ir1-Ir4 were synthesized and characterized. Possessing high singlet oxygen production ability and specific mitochondria-localization, Ir1 was developed as a mitochondria-targeting photosensitizer. Ir1 exhibited strong phototoxicity against cancer cell line A549 and its corresponding cisplatin-resistant one A549R. In contrast, Ir1 showed low cytotoxicity toward normal cell HLF. This selectivity resulted from the different uptake amount. With 405 nm irradiation, Ir1 induced mitochondria-mediated cell death in A549R cells, achieving the overcome of drug-resistant.
Collapse
Affiliation(s)
- Kai Xiong
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Ying Zhou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Xinlin Lin
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Junfeng Kou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Mingwei Lin
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Ruilin Guan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
26
|
Qiao H, Zhang L, Fang D, Zhu Z, He W, Hu L, Di L, Guo Z, Wang X. Surmounting tumor resistance to metallodrugs by co-loading a metal complex and siRNA in nanoparticles. Chem Sci 2021; 12:4547-4556. [PMID: 34163720 PMCID: PMC8179575 DOI: 10.1039/d0sc06680j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Copper complexes are promising anticancer agents widely studied to overcome tumor resistance to metal-based anticancer drugs. Nevertheless, copper complexes per se encounter drug resistance from time to time. Adenosine-5'-triphosphate (ATP)-responsive nanoparticles containing a copper complex CTND and B-cell lymphoma 2 (Bcl-2) small interfering RNA (siRNA) were constructed to cope with the resistance of cancer cells to the complex. CTND and siRNA can be released from the nanoparticles in cancer cells upon reacting with intracellular ATP. The resistance of B16F10 melanoma cells to CTND was terminated by silencing the cellular Bcl-2 gene via RNA interference, and the therapeutic efficacy was significantly enhanced. The nanoparticles triggered a cellular autophagy that amplified the apoptotic signals, thus revealing a novel mechanism for antagonizing the resistance of copper complexes. In view of the extensive association of Bcl-2 protein with cancer resistance to chemotherapeutics, this strategy may be universally applicable for overcoming the ubiquitous drug resistance to metallodrugs.
Collapse
Affiliation(s)
- Hongzhi Qiao
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China .,Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Lei Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Dong Fang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Zhenzhu Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 China
| | - Weijiang He
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Liuqing Di
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University Nanjing 210023 China
| |
Collapse
|
27
|
Li X, Zhao Y, Zhang T, Xing D. Mitochondria-Specific Agents for Photodynamic Cancer Therapy: A Key Determinant to Boost the Efficacy. Adv Healthc Mater 2021; 10:e2001240. [PMID: 33236531 DOI: 10.1002/adhm.202001240] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/02/2020] [Indexed: 02/06/2023]
Abstract
Mitochondria-targeted photodynamic therapy (Mt-PDT), which enables the photogenerated cytotoxic oxygen species with fatal oxidative damage to block mitochondrial functions, has been considered as a promising method to enhance the anticancer effectiveness. Aiming at the challenges of PDT, in the past few decades, numerous mitochondria-targeting molecular agents have been developed to boost the PDT efficacy via directly destroying the mitochondria or activating mitochondria-mediated cell death pathways. Herein, a review for recent advances of Mt-PDT is highlighted including: mitochondrial targeting design principles and strategies, therapeutic performance of mitochondria-targeted agents-mediated PDT as well as the agent-free Mt-PDT. In addition, it puts together the achievements of the combinatory mitochondria-anchoring PDT and other anticancer strategies, demonstrating the advantages provided by Mt-PDT. The existing challenges are discussed and future settlements for the development of mitochondria-specific agents are also forecasted.
Collapse
Affiliation(s)
- Xipeng Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
| | - Yu Zhao
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
| | - Tao Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
| |
Collapse
|
28
|
Cai DH, Zhang CL, Liu QY, He L, Liu YJ, Xiong YH, Le XY. Synthesis, DNA binding, antibacterial and anticancer properties of two novel water-soluble copper(II) complexes containing gluconate. Eur J Med Chem 2021; 213:113182. [PMID: 33486198 DOI: 10.1016/j.ejmech.2021.113182] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/02/2021] [Accepted: 01/07/2021] [Indexed: 12/20/2022]
Abstract
In this paper, two new Cu(II) complexes, [Cu(Gluc)(HPB)(H2O)]Gluc (CuG1) and [Cu(Gluc)(HPBC)(H2O)]Gluc (CuG2) (where HPB = 2-(2'-pyridyl)benzimidazole, HPBC = 5-chloro-2-(2'-pyridyl)benzimidazole, Gluc = d-Gluconic acid), with good water solubility were synthesized and characterized. These complexes exhibited a five-coordinated tetragonal pyramidal geometry. The DNA binding and cleavage properties of the complexes were investigated using multi-spectroscopy, viscosity measurement, molecular docking and gel electrophoresis analysis methods. The results showed that the complexes could interact with DNA by insertion and groove binding, and cleave CT-DNA through a singlet oxygen-dependent pathway in the presence of ascorbic acid. The studies on antibacterial and anticancer activities in vitro demonstrated that both complexes had good inhibitory activity against three Gram-positive bacteria (Staphylococcus aureus, Bacillus subtilis, Listeria monocytogenes) and one Gram-negative bacterium (Escherichia coli) and good cytotoxic activity toward the tested cancer cells (A549, HeLa and SGC-7901). CuG2 showed higher antimicrobial and cytotoxic activities than CuG1, which was consistent with their binding strength and cleavage ability to DNA, indicating that their antimicrobial and cytotoxic activities may be related to the DNA interaction. Moreover, the cell-based mechanism studies have indicated that CuG1 and CuG2 could arrest the cell cycle at G2/M phase, elevate the levels of intracellular reactive oxygen species (ROS) and decrease the mitochondrial membrane potential (MMP). The results showed that the complexes could induce apoptosis through DNA-damaged and ROS-mediated mitochondrial dysfunction pathways. Finally, the in vivo antitumor study revealed that CuG2 inhibited tumor growth by 50.44%, which is better than that of cisplatin (40.94%).
Collapse
Affiliation(s)
- Dai-Hong Cai
- Department of Applied Chemistry, South China Agricultural University, Guangzhou, 510642, PR China
| | - Chun-Lian Zhang
- Department of Applied Chemistry, South China Agricultural University, Guangzhou, 510642, PR China
| | - Qi-Yan Liu
- Department of Applied Chemistry, South China Agricultural University, Guangzhou, 510642, PR China
| | - Liang He
- Department of Applied Chemistry, South China Agricultural University, Guangzhou, 510642, PR China.
| | - Yun-Jun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Ya-Hong Xiong
- Department of Applied Chemistry, South China Agricultural University, Guangzhou, 510642, PR China
| | - Xue-Yi Le
- Department of Applied Chemistry, South China Agricultural University, Guangzhou, 510642, PR China.
| |
Collapse
|
29
|
Paul A, Singh P, Kuznetsov ML, Karmakar A, Guedes da Silva MFC, Koch B, Pombeiro AJL. Influence of anchoring moieties on new benzimidazole-based Schiff base copper(II) complexes towards estrogen dependent breast cancer cells. Dalton Trans 2021; 50:3701-3716. [PMID: 33634805 DOI: 10.1039/d0dt03873c] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Two new benzimidazole Schiff base copper(ii) compounds [Cu(5-CH2PPh3-2-salmethylben)(NO3)(H2O)][BF4]·2/3(H2O)·1/3(MeOH) (1) and [Cu(5-CH2NEt3-2-salmethylben)(Cl)][BF4] (2) were synthesised by mixing 2-(1-methyl-1H-benzo[d]imidazol-2-yl)aniline, (3-formyl-4-hydroxybenzyl)triphenylphosphonium chloride or N,N-diethyl-N-(3-formyl-4-hydroxybenzyl)ethanaminium chloride and Cu(NO3)2·3H2O or CuCl2·2H2O in the presence of tetrafluoroborate in a binary mixture of MeOH : H2O under refluxing conditions. The structures of the compounds were established by elemental analysis, FT-IR, ESI-MS analytical techniques and, for 1, by single-crystal X-ray diffraction analysis. Absorption and fluorescence spectroscopic methods were performed to evaluate the calf thymus DNA interactions with the compounds. The calculated binding constants (Kb) of 3.14 × 105 M-1 for 1 and 3.20 × 105 M-1 for 2 were established. The intercalative DNA binding mode was also verified by molecular docking studies. Both compounds demonstrated a notable in vitro cytotoxic effect against human A-549 (lung carcinoma), MCF-7 (breast cancer) and HeLa (cervical cancer) cancer cell lines. A substantial repressive effect on the proliferation of MCF-7 cells (breast cancer cells) was observed for compound 1. The mechanism of action for the effective antiproliferative activity of 1 has additionally been confirmed by means of various biological studies such as morphological assessment through AO/EB, detection of apoptotic induction via Hoechst/PI dual staining, flow cytometry for detection of cell cycle arrest, quantitative analysis of apoptotic cells, DNA degradation, generation of reactive oxygen species (ROS) and by apoptotic induction through mitochondrial staining.
Collapse
Affiliation(s)
- Anup Paul
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Priya Singh
- Departments of Zoology, Faculty of Science, Banaras Hindu University, Varanasi - 221 005, U.P., India.
| | - Maxim L Kuznetsov
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Anirban Karmakar
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - M Fátima C Guedes da Silva
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Biplob Koch
- Departments of Zoology, Faculty of Science, Banaras Hindu University, Varanasi - 221 005, U.P., India.
| | - Armando J L Pombeiro
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal. and Peoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| |
Collapse
|
30
|
The copper(II) complexes of new anthrahydrazone ligands: In vitro and in vivo antitumor activity and structure-activity relationship. J Inorg Biochem 2020; 212:111208. [DOI: 10.1016/j.jinorgbio.2020.111208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/23/2022]
|
31
|
Jin S, Muhammad N, Sun Y, Tan Y, Yuan H, Song D, Guo Z, Wang X. Multispecific Platinum(IV) Complex Deters Breast Cancer via Interposing Inflammation and Immunosuppression as an Inhibitor of COX‐2 and PD‐L1. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202011273] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Suxing Jin
- State Key Laboratory of Coordination Chemistry School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 P. R. China
| | - Nafees Muhammad
- State Key Laboratory of Coordination Chemistry School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 P. R. China
- School of Chemistry Sun Yat-Sen University Guangzhou 510275 P. R. China
| | - Yuewen Sun
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Yehong Tan
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| | - Hao Yuan
- State Key Laboratory of Coordination Chemistry School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 P. R. China
| | - Dongfan Song
- State Key Laboratory of Coordination Chemistry School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry School of Chemistry and Chemical Engineering Nanjing University Nanjing 210023 P. R. China
- Chemistry and Biomedicine Innovation Center Nanjing University Nanjing 210023 P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology School of Life Sciences Nanjing University Nanjing 210023 P. R. China
| |
Collapse
|
32
|
Jin S, Muhammad N, Sun Y, Tan Y, Yuan H, Song D, Guo Z, Wang X. Multispecific Platinum(IV) Complex Deters Breast Cancer via Interposing Inflammation and Immunosuppression as an Inhibitor of COX-2 and PD-L1. Angew Chem Int Ed Engl 2020; 59:23313-23321. [PMID: 32897000 DOI: 10.1002/anie.202011273] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/22/2022]
Abstract
Breast cancer (BC) is one of the most common malignancies in women and often accompanied by inflammatory processes. Cyclooxygenase-2 (COX-2) plays a vital role in the progression of BC, correlating with the expression of programmed death-ligand 1 (PD-L1). Overexpression of PD-L1 contributes to the immune escape of cancer cells, and its blockade would stimulate anticancer immunity. Two multispecific platinum(IV) complexes DNP and NP were prepared using non-steroidal antiinflammatory drug naproxen (NPX) as axial ligand(s) to inhibit the BC cells. DNP exhibited high cytotoxicity and antiinflammatory properties superior over NP, cisplatin and NPX; moreover, it displayed potent antitumor activity and almost no general toxicity in mice bearing triple-negative breast cancer (TNBC). Mechanistic studies revealed that DNP could downregulate the expression of COX-2 and PD-L1 in vitro and vivo, inhibit the secretion of prostaglandin, reduce the expression of BC-associated protein BRD4 and phosphorylation of extracellular signal-regulated kinases 1/2 (Erk1/2), and block the oncogene c-Myc in BC cells. These findings demonstrate that DNP is capable of intervening in inflammatory, immune, and metastatic processes of BC, thus presenting a new mechanism of action for anticancer platinum(IV) complexes. The multispecificity offers a special superiority for DNP to treat TNBC by combining chemotherapy and immunotherapy in one molecule.
Collapse
Affiliation(s)
- Suxing Jin
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Nafees Muhammad
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China.,School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Yuewen Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Yehong Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Hao Yuan
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Dongfan Song
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China.,Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
33
|
Novel copper complex CTB regulates methionine cycle induced TERT hypomethylation to promote HCC cells senescence via mitochondrial SLC25A26. Cell Death Dis 2020; 11:844. [PMID: 33041323 PMCID: PMC7548283 DOI: 10.1038/s41419-020-03048-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022]
Abstract
Related research has recognized the vital role of methionine cycle metabolism in cancers. However, the role and mechanism of methionine cycle metabolism in hepatocellular carcinoma are still unknown. In this study, we found that [Cu(ttpy-tpp)Br2]Br (Referred to as CTB) could induce hepatocellular carcinoma cells senescence, which is a new copper complex synthesized by our research group. Interestingly, CTB induces senescence by inhibiting the methionine cycle metabolism of HCC cells. Furthermore, the inhibitory effect of CTB on the methionine cycle depends on mitochondrial carrier protein SLC25A26, which was also required for CTB-induced HCC cells senescence. Importantly, we found that CTB-induced upregulation of SLC25A26 could cause abnormal methylation of TERT and inhibited TERT expression, which is considered to be an essential cause of cell senescence. The same results were also obtained in vivo, CTB inhibits the growth of subcutaneously implanted tumors in nude mice and promoted the expression of senescence markers in tumor tissues, and interference with SLC25A26 partially offset the antitumor effect of CTB.
Collapse
|
34
|
Schwarzbich S, Horstmann Née Gruschka C, Simon J, Siebe L, Moreth A, Wiegand C, Lavrentieva A, Scheper T, Stammler A, Bögge H, Fischer von Mollard G, Glaser T. Stronger Cytotoxicity for Cancer Cells Than for Fast Proliferating Human Stem Cells by Rationally Designed Dinuclear Complexes. Inorg Chem 2020; 59:14464-14477. [PMID: 32951424 DOI: 10.1021/acs.inorgchem.0c02255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cytostatic metallo-drugs mostly bind to the nucleobases of DNA. A new family of dinuclear transition metal complexes was rationally designed to selectively target the phosphate diesters of the DNA backbone by covalent bonding. The synthesis and characterization of the first dinuclear NiII2 complex of this family are presented, and its DNA binding and interference with DNA synthesis in polymerase chain reaction (PCR) are investigated and compared to those of the analogous CuII2 complex. The NiII2 complex also binds to DNA but forms fewer intermolecular DNA cross-links, while it interferes with DNA synthesis in PCR at lower concentrations than CuII2. To simulate possible competing phosphate-based ligands in vivo, these effects have been studied for both complexes with 100-200-fold excesses of phosphate and ATP, which provided no disturbance. The cytotoxicity of both complexes has been studied for human cancer cells and human stem cells with similar rates of proliferation. CuII2 shows the lowest IC50 values and a remarkable preference for killing the cancer cells. Three different assays show that the CuII2 complex induces apoptosis in cancer cells. These results are discussed to gain insight into the mechanisms of action and demonstrate the potential of this family of dinuclear complexes as anticancer drugs acting by a new binding target.
Collapse
Affiliation(s)
- Sabrina Schwarzbich
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Claudia Horstmann Née Gruschka
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Jasmin Simon
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Lena Siebe
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Alexander Moreth
- Lehrstuhl für Biochemie III, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Christiane Wiegand
- Lehrstuhl für Biochemie III, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Antonina Lavrentieva
- Zentrum Angewandte Chemie, Institut für Technische Chemie, Callinstrasse 5, D-30167 Hannover, Germany
| | - Thomas Scheper
- Zentrum Angewandte Chemie, Institut für Technische Chemie, Callinstrasse 5, D-30167 Hannover, Germany
| | - Anja Stammler
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Hartmut Bögge
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Gabriele Fischer von Mollard
- Lehrstuhl für Biochemie III, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| | - Thorsten Glaser
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstrasse 25, D-33615 Bielefeld, Germany
| |
Collapse
|
35
|
Kumar P, Butcher RJ, Patra AK. Ternary Co(II), Ni(II) and Cu(II) complexes containing dipyridophenazine and saccharin: Structures, reactivity, binding interactions with biomolecules and DNA damage activity. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2020.119532] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
36
|
Li S, Zhao J, Guo Y, Mei Y, Yuan B, Gan N, Zhang J, Hu J, Hou H. Influence of the introduction of a triphenylphosphine group on the anticancer activity of a copper complex. J Inorg Biochem 2020; 210:111102. [PMID: 32574870 DOI: 10.1016/j.jinorgbio.2020.111102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022]
Abstract
Aiming at obtaining new copper complexes with good cytotoxicity against cancer cells, triphenylphosphine (TPP) was introduced to obtain insight into the influence of the co-ligands. In this paper, two copper complexes, Cu(2-pbmq)(CH3OH)Br2 (1) and [Cu(2-pbmq)(TPP)Br]2 (2) were designed, synthesized, and characterized by X-ray crystallography, 2-((2-(pyrazin-2-yl)-1H-benzo[d]imidazol-1-yl)methyl))quinolone (2-pbmq), to investigate the influence of the TPP group on the anticancer activity of the metal complex. Although the presence of the TPP group diminished the intensity of the interaction properties of the complex with DNA, the in vitro anticancer activity and cellular uptake of the TPP-containing complex were markedly superior to those of its TPP-lacking counterpart. Detailed studies on the more potently cytotoxic complex 2 revealed that it accumulated in nucleus, arrested the cell cycle at the G0-G1 phase, causing mitochondrial dysfunction, involving the potential simultaneous mitochondrial membrane collapse, cellular ATP level depletion, and Ca2+ leakage, eventually inducing cell apoptosis. In summary, the introduction of a TPP group enhances the biological activity and cytotoxicity of the complex.
Collapse
Affiliation(s)
- Sen Li
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Jin'an Zhao
- College of Material and Chemical Engineering, Henan University of Urban Construction, Pingdingshan 467036, Henan, PR China.
| | - Yan Guo
- College of Material and Chemical Engineering, Henan University of Urban Construction, Pingdingshan 467036, Henan, PR China
| | - Yameng Mei
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Bangpeng Yuan
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Ning Gan
- College of Material and Chemical Engineering, Henan University of Urban Construction, Pingdingshan 467036, Henan, PR China
| | - Junshuai Zhang
- College of Material and Chemical Engineering, Henan University of Urban Construction, Pingdingshan 467036, Henan, PR China
| | - Jiyong Hu
- College of Material and Chemical Engineering, Henan University of Urban Construction, Pingdingshan 467036, Henan, PR China.
| | - Hongwei Hou
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| |
Collapse
|
37
|
Jyothi N, Ganji N, Daravath S, Shivaraj. Mononuclear cobalt(II), nickel(II) and copper(II) complexes: Synthesis, spectral characterization and interaction study with nucleotide by in vitro biochemical analysis. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.127799] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
|
39
|
Metal complexes for mitochondrial bioimaging. J Inorg Biochem 2020; 204:110985. [DOI: 10.1016/j.jinorgbio.2019.110985] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 02/07/2023]
|
40
|
Li S, Zhao J, Yuan B, Wang X, Zhang J, Yue L, Hou H, Hu J, Chen S. Crystal structure, DNA interaction and in vitro anticancer activity of Cu(II) and Pt(II) compounds based on benzimidazole-quinoline derivative. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
41
|
Li M, Shao J, Guo Z, Jin C, Wang L, Wang F, Jia Y, Zhu Z, Zhang Z, Zhang F, Zheng S, Wang X. Novel mitochondrion-targeting copper(II) complex induces HK2 malfunction and inhibits glycolysis via Drp1-mediating mitophagy in HCC. J Cell Mol Med 2020; 24:3091-3107. [PMID: 31994339 PMCID: PMC7077532 DOI: 10.1111/jcmm.14971] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 12/09/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023] Open
Abstract
[Cu(ttpy-tpp)Br2 ]Br (abbreviated as CTB) is a novel mitochondrion-targeting copper(II) complex synthesized by our research group, which contains tri-phenyl-phosphonium (TPP) groups as its lipophilic property. In this study, we explored how CTB affects mitochondrial functions and exerts its anti-tumour activity. Multiple functional and molecular analyses including Seahorse XF Bioanalyzer Platform, Western blot, immunofluorescence analysis, co-immunoprecipitation and transmission electron microscopy were used to elucidate the underlying mechanisms. Human hepatoma cells were subcutaneously injected into right armpit of male nude mice for evaluating the effects of CTB in vivo. We discovered that CTB inhibited aerobic glycolysis and cell acidification by impairing the activity of HK2 in hepatoma cells, accompanied by dissociation of HK2 from mitochondria. The modification of HK2 not only led to the complete dissipation of mitochondrial membrane potential (MMP) but also promoted the opening of mitochondrial permeability transition pore (mPTP), contributing to the activation of mitophagy. In addition, CTB co-ordinately promoted dynamin-related protein 1 (Drp1) recruitment in mitochondria to induce mitochondrial fission. Our findings established a previously unrecognized role for copper complex in aerobic glycolysis of tumour cells, revealing the interaction between mitochondrial HK2-mediated mitophagy and Drp1-regulated mitochondrial fission.
Collapse
Affiliation(s)
- Mengmeng Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pharmaceutical Technology, Xuzhou Pharmaceutical Vocational College, Xuzhou, China
| | - Jiangjuan Shao
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Chun Jin
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ling Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Jia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenzhu Zhu
- School of Food Science and Engineering, Nanjing University Of Finance & Economics, Nanjing, China
| | - Ziji Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
42
|
|
43
|
Shao J, Li M, Guo Z, Jin C, Zhang F, Ou C, Xie Y, Tan S, Wang Z, Zheng S, Wang X. TPP-related mitochondrial targeting copper (II) complex induces p53-dependent apoptosis in hepatoma cells through ROS-mediated activation of Drp1. Cell Commun Signal 2019; 17:149. [PMID: 31744518 PMCID: PMC6862763 DOI: 10.1186/s12964-019-0468-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/22/2019] [Indexed: 12/19/2022] Open
Abstract
Background In recent years, copper complexes have gradually become the focus of potential anticancer drugs due to their available redox properties and low toxicity. In this study, a novel mitochondrion-targeting copper (II) complex, [Cu (ttpy-tpp)Br2] Br (simplified as CTB), is first synthesized by our group. CTB with tri-phenyl-phosphine (TPP), a targeting and lipophilic group, can cross the cytoplasmic and mitochondrial membranes of tumor cells. The present study aims to investigate how CTB affects mitochondrial functions and exerts its anti-tumor activity in hepatoma cells. Methods Multiple molecular experiments including Flow cytometry, Western blot, Immunofluorescence, Tracker staining, Transmission Electron Microscopy and Molecular docking simulation were used to elucidate the underlying mechanisms. Human hepatoma cells were subcutaneously injected into right armpit of male nude mice for evaluating the effects of CTB in vivo. Results CTB induced apoptosis via collapse of mitochondrial membrane potential (MMP), ROS production, Bax mitochondrial aggregation as well as cytochrome c release, indicating that CTB-induced apoptosis was associated with mitochondrial pathway in human hepatoma cells. Mechanistic study revealed that ROS-related mitochondrial translocation of p53 was involved in CTB-mediated apoptosis. Simultaneously, elevated mitochondrial Drp1 levels were also observed, and interruption of Drp1 activation played critical role in p53-dependent apoptosis. CTB also strongly suppressed the growth of liver cancer xenografts in vivo. Conclusion In human hepatoma cells, CTB primarily induces mitochondrial dysfunction and promotes accumulation of ROS, leading to activation of Drp1. These stimulation signals accelerate mitochondrial accumulation of p53 and lead to the eventual apoptosis. Our research shows that CTB merits further evaluation as a chemotherapeutic agent for the treatment of Hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Mengmeng Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,Department of Pharmaceutical Technology, Xuzhou Pharmaceutical Vocational College, Xuzhou, 221116, China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Chun Jin
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunyan Ou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yaochen Xie
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shanzhong Tan
- The Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Zhenyi Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
44
|
Gao P, Pan W, Li N, Tang B. Boosting Cancer Therapy with Organelle-Targeted Nanomaterials. ACS APPLIED MATERIALS & INTERFACES 2019; 11:26529-26558. [PMID: 31136142 DOI: 10.1021/acsami.9b01370] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The ultimate goal of cancer therapy is to eliminate malignant tumors while causing no damage to normal tissues. In the past decades, numerous nanoagents have been employed for cancer treatment because of their unique properties over traditional molecular drugs. However, lack of selectivity and unwanted therapeutic outcomes have severely limited the therapeutic index of traditional nanodrugs. Recently, a series of nanomaterials that can accumulate in specific organelles (nucleus, mitochondrion, endoplasmic reticulum, lysosome, Golgi apparatus) within cancer cells have received increasing interest. These rationally designed nanoagents can either directly destroy the subcellular structures or effectively deliver drugs into the proper targets, which can further activate certain cell death pathways, enabling them to boost the therapeutic efficiency, lower drug dosage, reduce side effects, avoid multidrug resistance, and prevent recurrence. In this Review, the design principles, targeting strategies, therapeutic mechanisms, current challenges, and potential future directions of organelle-targeted nanomaterials will be introduced.
Collapse
Affiliation(s)
- Peng Gao
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| |
Collapse
|
45
|
Liu Y, Song X, Li S, Liu X, Tian J, Xu J, Yan S. Three pairs of enantiomers bearing mitochondria‐targeted TPP
+
groups as potential anti‐cancer agents. Appl Organomet Chem 2019. [DOI: 10.1002/aoc.4920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Yue Liu
- Department of ChemistryNankai University Tianjin 300071 People's Republic of China
- Key Laboratory of Advanced Energy Materials Chemistry (MOE) Tianjin 300071 People's Republic of China
| | - Xue‐Qing Song
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of PharmacyTianjin Medical University Tianjin 300070 China
| | - Si‐Tong Li
- Department of ChemistryNankai University Tianjin 300071 People's Republic of China
| | - Xin Liu
- Department of ChemistryNankai University Tianjin 300071 People's Republic of China
- Key Laboratory of Advanced Energy Materials Chemistry (MOE) Tianjin 300071 People's Republic of China
| | - Jin‐Lei Tian
- Department of ChemistryNankai University Tianjin 300071 People's Republic of China
- Key Laboratory of Advanced Energy Materials Chemistry (MOE) Tianjin 300071 People's Republic of China
| | - Jing‐Yuan Xu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of PharmacyTianjin Medical University Tianjin 300070 China
| | - Shi‐Ping Yan
- Department of ChemistryNankai University Tianjin 300071 People's Republic of China
- Key Laboratory of Advanced Energy Materials Chemistry (MOE) Tianjin 300071 People's Republic of China
| |
Collapse
|
46
|
Janković N, Trifunović Ristovski J, Vraneš M, Tot A, Petronijević J, Joksimović N, Stanojković T, Đorđić Crnogorac M, Petrović N, Boljević I, Matić IZ, Bogdanović GA, Mikov M, Bugarčić Z. Discovery of the Biginelli hybrids as novel caspase-9 activators in apoptotic machines: Lipophilicity, molecular docking study, influence on angiogenesis gene and miR-21 expression levels. Bioorg Chem 2019; 86:569-582. [DOI: 10.1016/j.bioorg.2019.02.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/03/2019] [Accepted: 02/09/2019] [Indexed: 02/06/2023]
|
47
|
Yi XQ, He YF, Cao YS, Shen WX, Lv YY. Porphyrinic Probe for Fluorescence "Turn-On" Monitoring of Cu + in Aqueous Buffer and Mitochondria. ACS Sens 2019; 4:856-864. [PMID: 30868875 DOI: 10.1021/acssensors.8b01240] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A zinc(II) porphyrin derivative (ZPSN) was designed and synthesized, and this probe exhibited rapid, selective and reversible binding to Cu+ for fluorescence monitoring in pure aqueous buffer. The detection mechanism is based on Cu+-activated disruption of axial coordination between the pyridyl ligand and the zinc center, which changes the molecular geometry and inhibits intramolecular electron transfer (ET), leading to fluorescence enhancement of the probe. The proposed sensing mechanism was supported by UV-vis spectroscopy/fluorescence spectral titration, NMR spectroscopy, mass spectrometry, and time-resolved fluorescence decay studies. The dissociation constant was calculated to be 6.53 × 10-11 M. CLSM analysis strongly suggested that ZPSN could penetrate live cells and successfully visualize Cu+ in mitochondria. This strategy may establish a design and offer a potential building block for construction of other metal sensors based on a similar mechanism.
Collapse
Affiliation(s)
- Xiao-Qin Yi
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, People’s Republic of China
- College of Pharmacy, Zhejiang University, Hangzhou, Zhejiang 310027, People’s Republic of China
| | - Yuan-Fan He
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, People’s Republic of China
| | - Yu-Sheng Cao
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, People’s Republic of China
| | - Wang-Xing Shen
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, People’s Republic of China
| | - Yuan-Yuan Lv
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, People’s Republic of China
| |
Collapse
|
48
|
Abstract
Background:
Since the serendipitous discovery of the antitumor activity of cisplatin
there has been a continuous surge in studies aimed at the development of new cytotoxic
metal complexes. While the majority of these complexes have been designed to interact with
nuclear DNA, other targets for anticancer metallodrugs attract increasing interest. In cancer
cells the mitochondrial metabolism is deregulated. Impaired apoptosis, insensitivity to antigrowth
signals and unlimited proliferation have been linked to mitochondrial dysfunction. It
is therefore not surprising that mitochondria have emerged as a major target for cancer therapy.
Mitochondria-targeting agents are able to bypass resistance mechanisms and to (re-) activate
cell-death programs.
Methods:
Web-based literature searching tools such as SciFinder were used to search for reports
on cytotoxic metal complexes that are taken up by the mitochondria and interact with
mitochondrial DNA or mitochondrial proteins, disrupt the mitochondrial membrane potential,
facilitate mitochondrial membrane permeabilization or activate mitochondria-dependent celldeath
signaling by unbalancing the cellular redox state. Included in the search were publications
investigating strategies to selectively accumulate metallodrugs in the mitochondria.
Results:
This review includes 241 references on antimitochondrial metal complexes, the use
of mitochondria-targeting carrier ligands and the formation of lipophilic cationic complexes.
Conclusion:
Recent developments in the design, cytotoxic potency, and mechanistic understanding
of antimitochondrial metal complexes, in particular of cyclometalated Au, Ru, Ir and
Pt complexes, Ru polypyridine complexes and Au-N-heterocyclic carbene and phosphine
complexes are summarized and discussed.
Collapse
Affiliation(s)
- Andrea Erxleben
- School of Chemistry, National University of Ireland, Galway, Ireland
| |
Collapse
|
49
|
Golbaghi G, Haghdoost MM, Yancu D, Santos YLDL, Doucet N, Patten SA, Sanderson JT, Castonguay A. Organoruthenium(II) Complexes Bearing an Aromatase Inhibitor: Synthesis, Characterization, in Vitro Biological Activity and in Vivo Toxicity in Zebrafish Embryos. Organometallics 2019; 38:702-711. [PMID: 31762529 DOI: 10.1021/acs.organomet.8b00897] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Third-generation aromatase inhibitors such as anastrozole (ATZ) and letrozole (LTZ) are widely used to treat estrogen receptor-positive ER+ breast cancers in postmenopausal women. Investigating their ability to coordinate metals could lead to the emergence of a new category of anticancer drug candidates with a broader spectrum of pharmacological activities. In this study, a series of ruthenium (II) arene complexes bearing the aromatase inhibitor anastrozole was synthesized and characterized. Among these complexes, [Ru(η 6 -C6H6)(PPh3)(η 1 -ATZ)Cl]BPh4 (3) was found to be the most stable in cell culture media, to lead to the highest cellular uptake and in vitro cytotoxicity in two ER+ human breast cancer cell lines (MCF7 and T47D), and to induce a decrease in aromatase activity in H295R cells. Exposure of zebrafish embryos to complex 3 (12.5 μM) did not lead to noticeable signs of toxicity over 96 h, making it a suitable candidate for further in vivo investigations.
Collapse
Affiliation(s)
- Golara Golbaghi
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Mohammad Mehdi Haghdoost
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Debbie Yancu
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Yossef López de Los Santos
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Nicolas Doucet
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Shunmoogum A Patten
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - J Thomas Sanderson
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Annie Castonguay
- INRS - Institut Armand-Frappier, Université du Québec, 531 boul. des Prairies, Laval, Québec, H7V 1B7, Canada
| |
Collapse
|
50
|
Zhao YP, Wang F, Jiang W, Liu J, Liu BL, Qi LW, Zhou W. A mitochondrion-targeting tanshinone IIA derivative attenuates myocardial hypoxia reoxygenation injury through a SDH-dependent antioxidant mechanism. J Drug Target 2019; 27:896-902. [DOI: 10.1080/1061186x.2019.1566338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yun-Peng Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Fei Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Wei Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Jinfeng Liu
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Bao-Lin Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Lian-Wen Qi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Wen Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| |
Collapse
|