Co-reporter:Kangqiang Qiu, Jinquan Wang, Cuilan Song, Lili Wang, Hongyi Zhu, Huaiyi Huang, Juanjuan Huang, Hui Wang, Liangnian Ji, and Hui Chao
ACS Applied Materials & Interfaces June 7, 2017 Volume 9(Issue 22) pp:18482-18482
Publication Date(Web):May 5, 2017
DOI:10.1021/acsami.7b02977
Synergistic photodynamic therapy (PDT) that combines photosensitizers (PSs) to attack different key sites in cancer cells is very attractive. However, the use of multiple PSs may increase dark cytotoxicity. Additionally, realizing the multiple vein passage of several PSs through dosing could be a challenge in clinical treatment. To address these issues, a novel strategy that enables a single PS to ablate two key sites (i.e., cytomembranes on the outside and mitochondria on the inside) of cancer cells synergistically was proposed. Five new fluorinated ruthenium (II) complexes (Ru1–Ru5), which possessed excellent two-photon properties and good singlet oxygen quantum yields, were designed and synthesized. When incubated with HeLa cells, the complexes were observed on the cytomembranes at first. With an extension of the treatment time, both the cytomembranes and mitochondria were lit up by the complexes. Under two-photon laser irradiation, the mitochondria and cytomembranes were ablated simultaneously, and the HeLa cells were destroyed effectively by the complexes, whether the cells were in a monolayer or in multicellular spheroids. With the largest phototoxicity index under the two-photon laser, Ru4 was used for two-photon PDT of in vivo xenograft tumors and successfully inhibited the growth of the tumors. Our results emphasized that the strategy of attacking two key sites with a single PS is an efficient method for PDT.Keywords: cytomembrane; fluorination; mitochondria; ruthenium (II) complex; Two-photon photodynamic therapy;
Co-reporter:Hui-juan Yu, Jiang-ping Liu, Zhi-feng Hao, Jun He, Ming Sun, Sheng Hu, Lin Yu, Hui Chao
Dyes and Pigments 2017 Volume 136() pp:416-426
Publication Date(Web):January 2017
DOI:10.1016/j.dyepig.2016.08.059
•Two new ruthenium complexes have been synthesized and characterized.•Complex 1 shows great photocytotoxicity towards Hela cells (IC50 = 1.9 μM).•Complex 1 inhibits Hela cell proliferation by induction of G2M phase cycle arrest and cells apoptosis.Two new Ru(II) complexes [Ru(dtzp)(dppz)Cl]+1 and [Ru(dtzp)(dppz)CH3CN]2+2 (dtzp = 2,6-di(thiazo1-2-yl)pyridine; dppz = dipyrido[3,2-a:2′,3′-c]phenazine) have been synthesized and evaluated as photodynamic anticancer agents. The results of the spectra titration, thermal denaturation and electrophoresis experiments suggest that both complexes could intercalatively bind to DNA and photocleave DNA efficiently by ROS generation and photoinduced electron transfer. When incubated under visible light (470 nm), complex 1 and 2 generate great photocytoxicity towards Hela cells in both 2D cancer cell monolayer and 3D MCTS cancer models, and a much greater photocytotoxicity was observed for complex 1, which may be associate with its' larger cellular uptake efficiency and stronger absorption at 470 nm. Flow cytometry analysis and immunofluorescence assay revealed that complex 1 inhibited Hela cell proliferation through G2M phase cycle arrest and cell apoptosis and could generate great photodamage to chromatin DNA. Complex 1 may be a prominent PDT candidate used for treating cervical carcinoma.
Co-reporter:Jinquan Wang, Junfeng Kou, Xiaojuan Hou, Zizhuo Zhao, Hui Chao
Inorganica Chimica Acta 2017 Volume 454() pp:176-183
Publication Date(Web):1 January 2017
DOI:10.1016/j.ica.2016.04.050
•A ruthenium(II) complex Ru-ipad can detect repeated cycles of hypoxia–normoxia.•Ru-ipad showed cytotoxicity against hypoxic 2D and 3D cancer cells.•Ru-ipad displayed hypoxia-targeted antitumor effects through inhibiting HIF-1α.Hypoxia is a common feature of most solid tumors and its precise imaging is of great importance to therapy planning. Here we demonstrated that a ruthenium(II) anthraquinone complex [Ru(bpy)2(ipad)](ClO4)2 (Ru-ipad, where bpy = 2,2′-bipyridine and ipad = 2-(anthracene-9,10-dione-2-yl)imidazo[4,5-f][1,10]phenanthroline), is a powerful optical probe that can detect repeated cycles of hypoxia–normoxia in living cells in real time. This hypoxia probe also showed cytotoxicity against hypoxic 2D cancer monolayer cultures and 3D multicellular tumor spheroids (MCTSs). The cytotoxicity of Ru-ipad was in accordance with its suppression of the hypoxia-inducible factor-1α (HIF-1α) protein in tumor cells. Therefore, Ru-ipad not only has the potential to detect hypoxia with high sensitivity but also displays hypoxia-targeted antitumor effects through the inhibition of HIF-1α expression. The discovery of this dual-function ruthenium(II) anthraquinone complex may represent an important advance in hypoxic solid tumor treatment.A ruthenium (II) anthraquinone complex Ru-ipad can not only detect repeated cycles of hypoxia–normoxia but also display hypoxia-targeted antitumor effects.
Co-reporter:Pingyu Zhang;Jinquan Wang;Huaiyi Huang;Kangqiang Qiu;Juanjuan Huang;Liangnian Ji
Journal of Materials Chemistry B 2017 vol. 5(Issue 4) pp:671-678
Publication Date(Web):2017/01/25
DOI:10.1039/C6TB01991A
Gold nanorods (AuNRs) and nanostars (AuNTs) were widely applied in photothermal cancer therapy recently. However, due to the photothermal effect, naked AuNRs and AuNTs easily melt into gold spheres. This drawback results in loss of the characteristic near-infrared (NIR) surface plasmon resonance (SPR) and limits their therapeutic applications. In this paper, we reported that ruthenium(II) complex-functionalized AuNRs (AuNRs@Ru) and AuNTs (AuNTs@Ru) exhibit higher photothermal stability and photothermal efficiency than naked AuNRs and AuNTs. AuNRs@Ru and AuNTs@Ru maintain the morphology and NIR SPR absorption of gold nanoparticles upon 0.25 W cm−2 laser irradiation, which is lower than the maximal permissible exposure of skin as per ANSI regulation (0.33 W cm−2 at 808 nm). Further photothermal therapy studies on three-dimensional (3D) HeLa spheroids and an in vivo tumor model show that AuNRs@Ru and AuNTs@Ru are more effective for the photothermal destruction of tumors than AuNRs and AuNTs.
Co-reporter:Leli Zeng;Shi Kuang;Guanying Li;Chengzhi Jin;Liangnian Ji
Chemical Communications 2017 vol. 53(Issue 12) pp:1977-1980
Publication Date(Web):2017/02/07
DOI:10.1039/C6CC10330H
A glutathione (GSH)-activatable ruthenium(II)-azo photosensitizer was prepared. The complex had low toxicity towards cells under dark conditions. It exhibited excellent phototoxicity under two-photon excitation (810 nm) and thus was developed as a two-photon photodynamic anticancer agent for cancer therapy.
Co-reporter:Chengzhi Jin;Ruilin Guan;Jingheng Wu;Bo Yuan;Lili Wang;Juanjuan Huang;Hui Wang;Liangnian Ji
Chemical Communications 2017 vol. 53(Issue 75) pp:10374-10377
Publication Date(Web):2017/09/19
DOI:10.1039/C7CC05193J
A series of NIR-emitting iridium(III) complexes were developed for multimodal phosphorescence imaging (NIR imaging, phosphorescence lifetime imaging and time-gated imaging) of mitochondria in living cells, 3D multicellular spheroids (MTCCs) and hippocampus slice under two-photon excitation.
Co-reporter:Jiangping Liu;Chengzhi Jin;Bo Yuan;Yu Chen;Xingguo Liu;Liangnian Ji
Chemical Communications 2017 vol. 53(Issue 71) pp:9878-9881
Publication Date(Web):2017/08/31
DOI:10.1039/C7CC05518H
Herein we present a series of DCA-Ir(III) co-drug complexes that preferentially accumulate in mitochondria and selectively cause cancer cell metabolic alterations and were found to act in synergy by sensitizing cancer cells for PDT to achieve cancer-specific enhanced two-photon PDT in the hypoxic environment of multicellular tumor spheroids.
Co-reporter:Pingyu Zhang;Yi Wang;Kangqiang Qiu;Zhiqian Zhao;Rentao Hu;Chuanxin He;Qianling Zhang
Chemical Communications 2017 vol. 53(Issue 91) pp:12341-12344
Publication Date(Web):2017/11/14
DOI:10.1039/C7CC07776A
A novel near infrared (NIR) phosphorescent osmium complex (Os1) was developed for lysosome tracking and photodynamic therapy. Owing to its NIR photophysical properties, cellular imaging ability and phototoxicity, it has advantages over its ruthenium analogue (Ru1).
Co-reporter:Huaiyi Huang, Nicolas Humbert, Vincent Bizet, Malay Patra, Hui Chao, Clément Mazet, Gilles Gasser
Journal of Organometallic Chemistry 2017 Volume 839(Volume 839) pp:
Publication Date(Web):15 June 2017
DOI:10.1016/j.jorganchem.2016.12.010
•The stability in DMF and DMSO of Ir(III) complexes is presented.•The cytotoxicity of Ir(III) complexes is described.•The role of the solvents used in biological studies is discussed.The stability of a compound in the solvent in which it is dissolved is a fundamental parameter in medicinal chemistry. In this article, we report on the results of our investigations on the stability of five Ir(III) complexes in DMF and DMSO. Importantly, we demonstrate that the rate of ligand exchange/decomposition of these compounds has an influence on their in vitro anticancer properties. The compounds were generally found to be less toxic to cancer cells after having been dissolved for longer time (24 h) in DMSO compared to short incubation time (1 h) in the same solvent. On the contrary, only minor differences in cytotoxicity were observed when the compounds were dissolved in DMF, emphasizing that this solvent should be employed instead of DMSO when unstable compounds are investigated, provided that the concentration of DMF is kept at a low concentration level.Download high-res image (111KB)Download full-size image
Co-reporter:Dr. Pingyu Zhang;Cookson K. C. Chiu;Dr. Huaiyi Huang;Yuko P. Y. Lam;Dr. Abraha Habtemariam;Thomas Malcomson; Martin J. Paterson;Dr. Guy J. Clarkson; Peter B. O'Connor; Hui Chao; Peter J. Sadler
Angewandte Chemie 2017 Volume 129(Issue 47) pp:14968-14968
Publication Date(Web):2017/11/20
DOI:10.1002/ange.201710973
Photodynamische Therapien bieten neue Möglichkeiten, um Krebszellen unschädlich zu machen und Resistenzen zu überwinden. In ihrer Zuschrift auf S. 15094 berichten P. B. O'Connor, H. Chao, P. J. Sadler et al. über lumineszierende Organoiridiumkomplexe mit langlebigen angeregten Zuständen, die Krebszellen nach Zwei-Photonen-Aktivierung mit rotem Licht selektiv schädigen. Die Komplexe erzeugen Sauerstoff, der wichtige zelluläre Proteine angreift. (Bildgestaltung: Juanjuan Huang)
Co-reporter:Dr. Pingyu Zhang;Cookson K. C. Chiu;Dr. Huaiyi Huang;Yuko P. Y. Lam;Dr. Abraha Habtemariam;Thomas Malcomson; Martin J. Paterson;Dr. Guy J. Clarkson; Peter B. O'Connor; Hui Chao; Peter J. Sadler
Angewandte Chemie International Edition 2017 Volume 56(Issue 47) pp:14898-14902
Publication Date(Web):2017/11/20
DOI:10.1002/anie.201709082
AbstractStrongly luminescent iridium(III) complexes, [Ir(C,N)2(S,S)]+ (1) and [Ir(C,N)2(O,O)] (2), containing C,N (phenylquinoline), O,O (diketonate), or S,S (dithione) chelating ligands, have been characterized by X-ray crystallography and DFT calculations. Their long phosphorescence lifetimes in living cancer cells give rise to high quantum yields for the generation of 1O2, with large 2-photon absorption cross-sections. 2 is nontoxic to cells, but potently cytotoxic to cancer cells upon brief irradiation with low doses of visible light, and potent at sub-micromolar doses towards 3D multicellular tumor spheroids with 2-photon red light. Photoactivation causes oxidative damage to specific histidine residues in the key proteins in aldose reductase and heat-shock protein-70 within living cancer cells. The oxidative stress induced by iridium photosensitizers during photoactivation can increase the levels of enzymes involved in the glycolytic pathway.
Co-reporter:Dr. Pingyu Zhang;Cookson K. C. Chiu;Dr. Huaiyi Huang;Yuko P. Y. Lam;Dr. Abraha Habtemariam;Thomas Malcomson; Martin J. Paterson;Dr. Guy J. Clarkson; Peter B. O'Connor; Hui Chao; Peter J. Sadler
Angewandte Chemie International Edition 2017 Volume 56(Issue 47) pp:14774-14774
Publication Date(Web):2017/11/20
DOI:10.1002/anie.201710973
Photodynamic therapy can be used to introduce novel mechanisms of killing cancer cells that overcome resistance. In their Communication on page 14898 ff., P. B. O'Connor, H. Chao, P. J. Sadler et al. describe luminescent organoiridium complexes, with long-lived excited states, which selectively damage cancer cells upon activation by two-photon red light, thus producing oxygen which attacks key cellular proteins (cover art by Juanjuan Huang).
Co-reporter:Dr. Pingyu Zhang;Cookson K. C. Chiu;Dr. Huaiyi Huang;Yuko P. Y. Lam;Dr. Abraha Habtemariam;Thomas Malcomson; Martin J. Paterson;Dr. Guy J. Clarkson; Peter B. O'Connor; Hui Chao; Peter J. Sadler
Angewandte Chemie 2017 Volume 129(Issue 47) pp:15094-15098
Publication Date(Web):2017/11/20
DOI:10.1002/ange.201709082
AbstractStrongly luminescent iridium(III) complexes, [Ir(C,N)2(S,S)]+ (1) and [Ir(C,N)2(O,O)] (2), containing C,N (phenylquinoline), O,O (diketonate), or S,S (dithione) chelating ligands, have been characterized by X-ray crystallography and DFT calculations. Their long phosphorescence lifetimes in living cancer cells give rise to high quantum yields for the generation of 1O2, with large 2-photon absorption cross-sections. 2 is nontoxic to cells, but potently cytotoxic to cancer cells upon brief irradiation with low doses of visible light, and potent at sub-micromolar doses towards 3D multicellular tumor spheroids with 2-photon red light. Photoactivation causes oxidative damage to specific histidine residues in the key proteins in aldose reductase and heat-shock protein-70 within living cancer cells. The oxidative stress induced by iridium photosensitizers during photoactivation can increase the levels of enzymes involved in the glycolytic pathway.
Co-reporter:Jiangping Liu;Chengzhi Jin;Bo Yuan;Xingguo Liu;Yu Chen;Liangnian Ji
Chemical Communications 2017 vol. 53(Issue 12) pp:2052-2055
Publication Date(Web):2017/02/07
DOI:10.1039/C6CC10015E
Herein a series of mitochondria-targeted AIE (aggregation-induced emission)-active Ir(III) complexes were designed to selectively exert one-/two-photon photodynamic activities in mitochondria to address the issues which current PDT are confronted with (i.e., shallow penetration depth of routinely used irradiation; systematic toxicity associated with effective drug concentration; concentration-quenched photodynamic activity at the target, etc.).
Co-reporter:Jin-Quan Wang;Jun-Feng Kou;Zi-Zhuo Zhao;Kang-Qiang Qiu
Inorganic Chemistry Frontiers 2017 vol. 4(Issue 6) pp:1003-1012
Publication Date(Web):2017/06/13
DOI:10.1039/C7QI00149E
Metastasis is a major health threat for most cancer patients, thus anti-metastasis treatments that reduce cell migration and invasion are critical for cancer treatment. In this study, four anthraquinone-bridged diruthenium(II) complexes, [(bpy)2Ru(L)Ru(bpy)2]4+ (Ru1, L = 1,4-bis(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)anthracene-9,10-dione; Ru2, L = 1,5-bis(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)anthracene-9,10-dione; Ru3, L = 2,6-bis(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)anthracene-9,10-dione; and Ru4, L = 2,7-bis(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)anthracene-9,10-dione) were synthesized and characterized. These Ru(II) complexes exhibited multi-targeted anti-metastatic properties against human hepatocarcinoma MHCC97-H cells that included the inhibition of migration and invasion. Further investigation of the intracellular mechanisms revealed that Ru(II) complexes suppressed the phosphorylation of ERK and AKT. Moreover, significant reduction of the extracellular and intracellular expression of the metastatic regulatory proteins MMP-2 and MMP-9 was also observed after Ru1–Ru4 treatment. In addition, these Ru(II) complexes negatively modulate the actin cytoskeleton by inhibiting Cdc42 protein expression, arresting the cells in the G2/M phase. The results indicate that these ruthenium(II) complexes have potential as drug candidates for anti-metastatic therapies.
Co-reporter:Kangqiang Qiu;Miao Ouyang;Yukang Liu;Huaiyi Huang;Chaofeng Liu;Yu Chen;Liangnian Ji
Journal of Materials Chemistry B 2017 vol. 5(Issue 27) pp:5488-5498
Publication Date(Web):2017/07/12
DOI:10.1039/C7TB00731K
By integrating targeting, imaging and treatment, organelle-targeted photodynamic therapy (PDT) has been reported to be an effective strategy for cancer therapy. However, targeting leads to the accumulation of photosensitizers (PSs) in the targeted organelles, which leads to a reduction in 1O2 generation and fluorescence quenching, especially for the lipophilic mitochondria-targeted PSs. Moreover, because PSs always need exposure to light for a specific period, photobleaching is difficult to avoid. To address these issues, two iridium(III) complexes with aggregation-induced two-photon emission (AITPE) characteristics were developed. With lipophilicity, the complexes aggregated in water and targeted mitochondria. Owing to their impressive 1O2 production quantum yields and excellent two-photon properties in the aggregate states, the complexes were successfully used for mitochondria-targeted two-photon PDT in monolayer cells and multicellular spheroids. Our results highlighted that the use of a PS with aggregation enhanced 1O2 generation and fluorescence is an effective solution for aggregation in organelle-targeted PDT.
Co-reporter:Miao Ouyang;Leli Zeng;Huaiyi Huang;Chengzhi Jin;Jiangping Liu;Yu Chen;Liangnian Ji
Dalton Transactions 2017 vol. 46(Issue 20) pp:6734-6744
Publication Date(Web):2017/05/23
DOI:10.1039/C7DT01043E
Six cyclometalated iridium(III) complexes bearing different numbers of fluorine atoms were synthesized. These complexes demonstrated much better anti-proliferation activities towards five tumour cell lines than the widely used clinical chemotherapeutic agent cisplatin. Moreover, the anti-proliferation activities were correlated to the number of substituted fluorine atoms. Colocalization and inductively coupled plasma-mass spectrometry (ICP-MS) indicated that this series of complexes could penetrate cell membranes rapidly and preferentially target mitochondria. Manifesting high selectivity between tumour cells and normal cells and remarkable sensitivity to a cisplatin-resistant cell line (A549R), complex Ir6 was successfully developed as a novel anticancer agent (with IC50 values of 0.5 ± 0.1 μM for HeLa, 1.1 ± 0.2 μM for HepG2, 1.5 ± 0.3 μM for BEL-7402, 0.8 ± 0.1 μM for A549, and 0.7 ± 0.2 μM for A549R cell lines). Further mechanism studies including mitochondrial membrane potential depolarization and caspase 3/7 activation revealed that Ir6 induced apoptosis via mitochondrial pathways. These results demonstrated that complex Ir6 might be a promising candidate as a mitochondria-targeted theranostic anticancer agent.
Co-reporter:Kangqiang Qiu, Huaiyi Huang, Bingyang Liu, Yukang Liu, Ziyi Huang, Yu Chen, Liangnian Ji, and Hui Chao
ACS Applied Materials & Interfaces 2016 Volume 8(Issue 20) pp:12702-12710
Publication Date(Web):May 6, 2016
DOI:10.1021/acsami.6b03422
Lysosomes are the stomachs of the cells that degrade endocytosis and intracellular biomacromolecules and participate in various other cellular processes, such as apoptosis and cell migration. The ability of long-term tracking of lysosomes is very important to understand the details of lysosomal functions and to evaluate drug and gene delivery systems. For studying lysosomes, we designed and synthesized a water-soluble triscyclometalated iridium(III) complex (Ir-lyso) attaching morpholine moieties. The phosphorescent intensity of Ir-lyso is responsive to pH and decreases with an increase in the pH but not quenching in high pH. With excellent two-photon properties, Ir-lyso was used to light up the lysosomes in living cells and 3D tumor spheroids. Moreover, Ir-lyso could label lysosomes more than 4 days, so we developed this complex to act as a long-term probe for tracking lysosomes during cell migration and apoptosis. To the best of our knowledge, this is the first paradigm of metal complexes as the two-photon phosphorescent probe for long-term lysosomes tracking.
Co-reporter:Huaiyi Huang, Pingyu Zhang, Yu Chen, Kangqiang Qiu, Chengzhi Jin, Liangnian Ji and Hui Chao
Dalton Transactions 2016 vol. 45(Issue 33) pp:13135-13145
Publication Date(Web):25 May 2016
DOI:10.1039/C6DT01270A
DNA binding and DNA transcription inhibition is regarded as a promising strategy for cancer chemotherapy. Herein, chloro terpyridyl Ru(II) complexes, [Ru(tpy)(N^N)Cl]+ (Ru1, N^N = 2,2′-bipyridine; Ru2, N^N = 3-(pyrazin-2-yl)-as-triazino[5,6-f]acenaphthylene; Ru3, N^N = 3-(pyrazin-2-yl)-as-triazino[5,6-f]phenanthrene; Ru4, N^N = 3-(pyrazin-2-yl)-as-triazino[5,6-f]pyrene) were prepared as DNA intercalative and covalent binding anticancer agents. The chloro ligand hydrolysis slowly and the octanol and water partition coefficient of Ru2–Ru4 were between 0.6 and 1.2. MALDI-TOF mass, DNA gel electrophoresis confirmed covalent and intercalative DNA binding modes of Ru2–Ru4, while Ru1 can only bind DNA covalently. As a result, Ru2–Ru4 exhibited stronger DNA transcription inhibition activity, higher cell uptake efficiency and better anticancer activity than Ru1. Ru4 was the most toxic complex toward all cancer cells which inhibited DNA replication and transcription. AO/EB, Annexin V/PI, nuclear staining, JC-1 assays further confirmed that Ru2–Ru4 induced cancer cell death by an apoptosis mechanism.
Co-reporter:Chen Qian, Jingheng Wu, Liangnian Ji and Hui Chao
Dalton Transactions 2016 vol. 45(Issue 26) pp:10546-10555
Publication Date(Web):13 May 2016
DOI:10.1039/C6DT01422D
Four chiral Ru(II) complexes bearing furan ligands, Δ/Λ-[Ru(bpy)2(pocl)]2+ (Δ/Λ-1) and Δ/Λ-[Ru(bpy)2(poi)]2+ (Δ/Λ-2) (bpy = 2,2′-bipyridine, pocl = 2-(5-chlorofuran-2-yl)imidazo[4,5-f][1,10]phenanthroline, poi = 2-(5-5-iodofuran-2-yl)imidazo[4,5-f][1,10]phenanthroline), were synthesized and characterized. These Ru(II) complexes showed antitumor activities against HeLa, A549, HepG2, HL-60 and K562 tumor cell lines, especially the HL-60 tumor cell line. Moreover, Δ-2 was more active than other complexes accounting for the different cellular uptakes. In addition, Δ-2 could accumulate in the nucleus of HL-60 cells, suggesting that nucleic acids were the cellular target of Δ-2. Topoisomerase inhibition tests in vitro and in living cells confirmed that the four complexes acted as efficient topoisomerase IIα poisons, DNA double-strand breaks had also been observed from neutral single cell gel electrophoresis (comet assay). Δ-2 inhibited the growth of HL-60 cells through the induction of apoptotic cell death, as evidenced by the Alexa Fluor® 488 annexin V staining assays. The results demonstrated that Δ-2 acted as a topoisomerase IIα poison and caused DNA double-strand damage that could lead to apoptosis.
Co-reporter:Guanying Li, Lingli Sun, Liangnian Ji and Hui Chao
Dalton Transactions 2016 vol. 45(Issue 34) pp:13261-13276
Publication Date(Web):11 Jul 2016
DOI:10.1039/C6DT01624C
The DNA photoswitch [Ru(bpy)2dppz]2+ (bpy = 2,2′-bipyridine, dppz = dipyrido[3,2-a:2′,3′-c]phenazine) has attracted much attention and become a powerful tool for studying the interaction of metal polypyridyl complexes with DNA. A large number of Ru-dppz complexes have been designed for a wide range of uses in many fields. In this perspective, we first introduce the latest results of Ru-dppz complexes that bind with DNA. The mechanisms of the light-switch effect and the structural modifications of Ru-dppz systems are also briefly introduced. We also review the recent advances in biological applications of the Ru-dppz system in DNA binders, cellular imaging, anticancer drugs, protein aggregation detection and chemosensors.
Co-reporter:Li Xu, Yu-Ying Liu, Liu-Ming Chen, Ye-Yi Xie, Jie-Xing Liang, Hui Chao
Journal of Inorganic Biochemistry 2016 Volume 159() pp:82-88
Publication Date(Web):June 2016
DOI:10.1016/j.jinorgbio.2016.02.028
•A Ruthenium(II) complex with benzofuran group was synthesized.•A Ru(II) complex was utilized as a mitochondrial fluorescence probe.•The complex exhibited superior photostability compared with the commercialized dyes.Labeling and imaging mitochondria have attracted considerable interest because of its involvement in early stage apoptosis and necrotic cell death. Various highly specific and photostable fluorescent probes for mitochondria are in demand. In the present study, two novel Ru(II) polypyridine complexes Ru1 and Ru2 were developed to act as mitochondrial fluorescence probes. In comparison with the commercially available mitochondrial trackers, Ru1 possesses high mitochondria-specificity, superior photostability, high resistance to the loss of mitochondrial membrane potential and appreciable tolerance to environmental change, allowing imaging of the mitochondrial morphological changes over long periods of time. Combined results indicate that Ru1 may contribute to the future development of staining agents for organelle-selective imaging in living cells.A Ruthenium(II) complex with benzofuran group was found to possess a superior capacity to selectively accumulate in mitochondria, superior photostability, low cytotoxicity at the imaging concentration, high resistance to the loss of mitochondrial membrane potential and good water solubility. Thus, this complex demonstrates good potential in mitochondrial imaging in living cells.
Co-reporter:Lingli Sun;Yu Chen;Shi Kuang;Guanying Li;Ruilin Guan;Jiangping Liu; Liangnian Ji ;Dr. Hui Chao
Chemistry - A European Journal 2016 Volume 22( Issue 26) pp:8955-8965
Publication Date(Web):
DOI:10.1002/chem.201600310
Abstract
In the present study, four mitochondria-specific and two-photon phosphorescence iridium(III) complexes, Ir1–Ir4, were developed for mitochondria imaging in hypoxic tumor cells. The iridium(III) complex has two anthraquinone groups that are hypoxia-sensitive moieties. The phosphorescence of the iridium(III) complex was quenched by the functions of the intramolecular quinone unit, and it was restored through two-electron bioreduction under hypoxia. When the probes were reduced by reductase to hydroquinone derivative products under hypoxia, a significant enhancement in phosphorescence intensity was observed under one- (λ=405 nm) and two-photon (λ=720 nm) excitation, with a two-photon absorption cross section of 76–153 GM at λ=720 nm. More importantly, these probes possessed excellent specificity for mitochondria, which allowed imaging and tracking of the mitochondrial morphological changes in a hypoxic environment over a long period of time. Moreover, the probes can visualize hypoxic mitochondria in 3D multicellular spheroids and living zebrafish through two-photon phosphorescence imaging.
Co-reporter:Kangqiang Qiu, Huaiyi Huang, Bingyang Liu, Yukang Liu, Pingyu Zhang, Yu Chen, Liangnian Ji and Hui Chao
Journal of Materials Chemistry A 2015 vol. 3(Issue 32) pp:6690-6697
Publication Date(Web):20 Jul 2015
DOI:10.1039/C5TB01091H
Two-photon phosphorescent probes have emerged as promising molecular tools for imaging subcellular organelles. Here, the facile synthesis of four new iridium(III)-based mitochondrial probes with two-photon phosphorescence, Ir1–Ir4, is presented. Ir1–Ir4 possess high specificity for mitochondrial localization, which is advantageous in comparison with commercially available mitochondrial trackers of changes in the mitochondrial membrane potential in live cells. In addition to low cytotoxicity and high resistance to photobleaching, Ir1–Ir4 are applicable for imaging and tracking of mitochondrial morphological changes during the early stages of apoptosis. While naturally possessing intensive two-photon properties, Ir1–Ir4 were further developed for imaging of the mitochondria in 3D multicellular spheroids.
Co-reporter:Yu Chen, Wenchao Xu, Jiarui Zuo, Liangnian Ji and Hui Chao
Journal of Materials Chemistry A 2015 vol. 3(Issue 16) pp:3306-3314
Publication Date(Web):03 Mar 2015
DOI:10.1039/C5TB00251F
Mitochondrial dynamic processes are essential for mammalian development and are engaged in several diseases. However, commercial mitochondrial probes are currently too limited to satisfactorily track these dynamics. In this present work, seven dinuclear iridium(III) complexes [(ppy)2Ir(L1–7)Ir(ppy)2]2+ (Ir1–Ir7, ppy = 2-phenylpyridine, L = 1,3-bis(1-substituted-imidazo[4,5-f][1,10]phenanthroline-2-yl)benzene) were synthesized. Possessing a high specificity for mitochondria, low cytotoxicity and high resistance to the loss of the mitochondrial membrane potential, Ir2–Ir7 can serve as mitochondrial imaging dyes. Ir3 and Ir6 are further developed for tracking mitochondrial morphological changes during the early stages of apoptosis. In addition, the relationship of the electron donating/withdrawing groups in these systems with the photophysical properties and photostability is also discussed.
Co-reporter:Pingyu Zhang, Huaiyi Huang, Juanjuan Huang, Hongmin Chen, Jinquan Wang, Kangqiang Qiu, Donglei Zhao, Liangnian Ji, and Hui Chao
ACS Applied Materials & Interfaces 2015 Volume 7(Issue 41) pp:23278
Publication Date(Web):October 2, 2015
DOI:10.1021/acsami.5b07510
To enhance the efficacy and optimize the treatment of cancers, the integration of multimodal treatment strategies leading to synergistic effects is a promising approach. The coassembly of multifunctional agents for systematic therapies has received considerable interest in cancer treatment. Herein, Ru(II) complex-functionalized single-walled carbon nanotubes (Ru@SWCNTs) are developed as nanotemplates for bimodal photothermal and two-photon photodynamic therapy (PTT-TPPDT). SWCNTs have the ability to load a great amount of Ru(II) complexes (Ru1 or Ru2) via noncovalent π–π interactions. The loaded Ru(II) complexes are efficiently released by the photothermal effect of irradiation from an 808 nm diode laser (0.25 W/cm2). The released Ru(II) complexes produce singlet oxygen species (1O2) upon two-photon laser irradiation (808 nm, 0.25 W/cm2) and can be used as a two-photon photodynamic therapy (TPPDT) agent. Based on the combination of photothermal therapy and two-photon photodynamic therapy, Ru@SWCNTs have greater anticancer efficacies than either PDT using Ru(II) complexes or PTT using SWCNTs in two-dimensional (2D) cancer cell and three-dimensional (3D) multicellular tumor spheroid (MCTS) models. Furthermore, in vivo tumor ablation is achieved with excellent treatment efficacy under a diode laser (808 nm) irradiation at the power density of 0.25 W/cm2 for 5 min. This study examines an efficacious bimodal PTT and TPPDT nanoplat form for the development of cancer therapeutics.Keywords: photodynamic therapy; photothermal therapy; Ru(II) complex; single-walled carbon nanotubes; two-photon
Co-reporter:Bole Yu, Ying Chen, Mei Hong, Pingping Duan, Shifeng Gan, Hui Chao, Zujin Zhao and Jing Zhao
Chemical Communications 2015 vol. 51(Issue 76) pp:14365-14368
Publication Date(Web):03 Aug 2015
DOI:10.1039/C5CC05239D
Using an internally oxidizing directing group (DG) strategy, we report a RhIII-catalyzed synthesis of isoquinolones via C–H activation/annulation of benzoylhydrazines and alkynes. Tunable double cascade cyclization of benzoylhydrazines with two equivalents of alkynes led to tetracyclic amides. These N-heterocycles demonstrated adjustable AIE properties.
Co-reporter:Chengzhi Jin, Jiangping Liu, Yu Chen, Guanying Li, Ruilin Guan, Pingyu Zhang, Liangnian Ji and Hui Chao
Dalton Transactions 2015 vol. 44(Issue 16) pp:7538-7547
Publication Date(Web):18 Mar 2015
DOI:10.1039/C5DT00467E
A new series of cyclometalated iridium(III) complexes with imidazo[4,5-f][1,10]phenanthroline derivatives (i.e., MitoIr1–MitoIr7) were synthesized and developed to image mitochondria in living cells. In comparison with commercially available mitochondrial trackers, these complexes exhibit a superior capacity to selectively accumulate in mitochondria with no requirement of any membrane permeabilization or replacement of the culture medium. In addition, the excellent photostability under continuous laser irradiation as well as the stable physiological pH resistance of these complexes were confirmed by photobleaching experiments and luminescence measurements. Importantly, MitoIr7, which exhibited both excellent luminescence and high ability to locate in mitochondria, was developed to track the mitochondrial morphological changes over a long period of time.
Co-reporter:Huaiyi Huang, Pingyu Zhang, Yu Chen, Liangnian Ji and Hui Chao
Dalton Transactions 2015 vol. 44(Issue 35) pp:15602-15610
Publication Date(Web):23 Jul 2015
DOI:10.1039/C5DT02446C
Ruthenium complexes have been considered as promising substitutes for cisplatin in cancer chemotherapy. However, novel ruthenium-based therapies are faced with some limitations, such as unimpressive cytotoxicity toward solid tumors. Herein, we designed and synthesized phenyl-substituted terpyridyl ruthenium(II) complexes ([Ru(tpy)(bpy)Cl]+ (Ru1), [Ru(phtpy)(bpy)Cl]+ (Ru2) and [Ru(biphtpy)(bpy)Cl]+ (Ru3)) which exhibited distinctly different anticancer activity. Ru1–Ru3 all underwent moderate aquation in buffer solution and this process was significantly inhibited by high chloride concentration. Cancer cells were found to readily uptake the relatively hydrophobic Ru3, as quantified using inductively coupled plasma mass spectrometry (ICP-MS). Ru1 was found to be non-cytotoxic (IC50 > 100 μM) while Ru3 exhibited very promising cytotoxicity on both two-dimensional (2D) cancer cell monolayers and 3D MCTSs. An antiproliferative assay revealed that Ru3 significantly inhibited cellular DNA replication which ultimately induced apoptosis of cancer cells.
Co-reporter:Guoliang Liao, Xiang Chen, Jingheng Wu, Chen Qian, Yi Wang, Liangnian Ji and Hui Chao
Dalton Transactions 2015 vol. 44(Issue 34) pp:15145-15156
Publication Date(Web):07 Jan 2015
DOI:10.1039/C4DT03585B
One novel ruthenium polypyridyl complex, [Ru(bpy)2(icip)]2+ (1), and two previously reported ruthenium polypyridyl complexes, [Ru(bpy)2(pdppz)]2+ (2) and [Ru(bpy)2(tactp)]2+ (3) (bpy = 2,2′-bipyridine, icip = 2-(indeno[2,1-b]chromen-6-yl)-1H-imidazo[4,5-f][1,10]phenanthroline, pdppz = phenanthro[4,5-abc]dipyrido[3,2-h:2′,3′-j]phenazine, tactp = 4,5,9,18-tetraazachryseno[9,10-b]-triphenylene), have been synthesised. As expected, these complexes show inhibition towards telomerase by inducing and stabilising the G-quadruplex structure, and behave as topoisomerase I/II poisons at the same time. Additionally, the acute and chronic cytotoxicities of the complexes are considered. Furthermore, cell apoptosis experiments are used to briefly study the mechanism. Because studies involving multi-target inhibition towards topoisomerase and telomerase of Ru(II) complexes have not been reported previously, the present research may help to develop innovative chemical strategies and therapies.
Co-reporter:Huaiyi Huang, Pingyu Zhang, Bole Yu, Chengzhi Jin, Liangnian Ji and Hui Chao
Dalton Transactions 2015 vol. 44(Issue 39) pp:17335-17345
Publication Date(Web):01 Sep 2015
DOI:10.1039/C5DT02081F
This study investigated the photodynamic therapy (PDT) and anticancer activity of mixed ligand Ru(II) terpyridyl complexes (Ru1–Ru3). The photophysical and photochemical properties, hydrophobic properties, DNA binding and DNA transcription inhibition abilities, cell uptake efficiency, cellular localization and photo-cytotoxicity were investigated. Ru1–Ru3 exhibited red luminescence between 670–710 nm and functioned as photo-sensitizers (PSs) by generating both singlet oxygen and radical ions. Without light activation, Ru1–Ru3 were located at the cytoplasm and were nontoxic to cells. However, upon light activation, Ru1–Ru3 exhibited significant photocytotoxicity. After PDT treatment, mitochondria alteration and nuclear membrane disruption occurred, which resulted in relocalization of the complexes from the cytoplasm to the nucleus. Moreover, high cellular oxidative stress caused cell necrocytosis after PDT treatment.
Co-reporter:Kangqiang Qiu, Bole Yu, Huaiyi Huang, Pingyu Zhang, Liangnian Ji and Hui Chao
Dalton Transactions 2015 vol. 44(Issue 15) pp:7058-7065
Publication Date(Web):09 Mar 2015
DOI:10.1039/C5DT00117J
To prolong the observation time, increase the penetration depth and decrease self-absorption and phototoxicity, two-photon luminescent vectors have emerged as promising tools for tracking gene delivery in living cells. Herein, we report four new tetranuclear Ru(II) complexes based on oligo-oxyethylene and polybenzimidazole as one- and two- photon luminescent tracking non-viral gene vectors. In such a molecular design, the oligo-oxyethylene, polybenzimidazole and Ru(II) polypyridyl complexes were expected to render the vectors with increased cell permeability, biocompatibility, proton buffering capacity and one- and two-photon luminescence. Corresponding DNA interaction studies showed that the ability of the complexes to condense DNA decreased with increasing oligo-oxyethylene lengths. Additionally, all complexes protected DNA. The complexes were investigated as one- and two-photon tracking non-viral gene vectors in living cells and showed proper cellular uptake, good luciferase expression and low cytotoxicity.
Co-reporter:Hui-juan Yu, Shu-mei Huang, Hui Chao, Liang-nian Ji
Journal of Inorganic Biochemistry 2015 Volume 149() pp:80-87
Publication Date(Web):August 2015
DOI:10.1016/j.jinorgbio.2015.02.020
•Two new ruthenium complexes have been synthesized and characterized.•The complexes could photocleave DNA under anaerobic condition.•The anaerobic photocleavage functions via photoinduced electron transfer pathway.Two new Ru(II) complexes [Ru(tpy)(dpoq)Cl]+1 and [Ru(tpy)(dpoq)CH3CN]2 +2 (tpy = 2,2':6',2''-terpyridine; dpoq = dipyrido[1,2,5]oxadiazolo[3,4-b]quinoxaline) have been synthesized and characterized by elemental analysis, 1H NMR, electrospray ionization mass spectra (ESI-MS) and X-ray crystallographic study. The experimental results of spectra titration, thermal denaturation and viscosity measurements suggest that the two complexes intercalatively bind to DNA. When irradiated under light, the two complexes could efficiently photocleave DNA both under aerobic and anaerobic condition. The mechanism studies reveal that the photocleavage reaction functions through both oxygen-independent (photoinduced electron transfer, type III reaction) and oxygen-dependent (singlet oxygen generation, type II reaction) pathways and the oxygen-independent pathway is the major process. These complexes will be more promising photodynamic therapy (PDT) candidates used for treating hypoxic tumors.The complexes could photocleave DNA both under aerobic and anaerobic condition. The photocleavage process was found to function through singlet oxygen generation and photoinduced electron transfer pathways.
Co-reporter:Jin-Quan Wang, Ping-Yu Zhang, Liang-Nian Ji, Hui Chao
Journal of Inorganic Biochemistry 2015 Volume 146() pp:89-96
Publication Date(Web):May 2015
DOI:10.1016/j.jinorgbio.2015.02.003
•The antitumor activity of Δ-Ru1 was evaluated.•The antitumor activity of Δ-Ru1 was comparable with cisplatin in vivo.•Δ-Ru1 exerts its cytotoxicity through the mitochondria-mediated apoptotic pathway.•Δ-Ru1 inhibits tumor growth in vivo with fewer side-effects than cisplatin.The antitumor activity of a ruthenium(II) polypyridyl complex, Δ-[Ru(bpy)2(HPIP)](ClO4)2 (Δ-Ru1, where bpy = 2,2′-bipyridine, HPIP = 2-(2-hydroxyphenyl)imidazo[4,5-f][1,10]phenanthroline), was evaluated. The in vivo experiments showed that Δ-Ru1 inhibited the growth of a human cervical carcinoma cell line (HeLa) xenotransplanted into nude mice with efficiency similar to that of cisplatin. Histopathology examination of the tumors from treated xenograft models was consistent with apoptosis in tumor cells. Importantly, in striking contrast with cisplatin, Δ-Ru1 did not cause any detectable side effects on the kidney, liver, peripheral neuronal system, or the hematological system at the pharmacologically effective dose. The preclinical studies reported here provide support for the clinical use of Δ-Ru1 as an exciting new drug candidate with lower toxicity than cisplatin, endowed with proapoptotic properties.A ruthenium(II) polypyridyl complex, Δ-[Ru(bpy)2(HPIP)](ClO4)2 (Δ-Ru1), was found to inhibit tumor growth in vivo without causing any detectable side effects on the kidney, liver, peripheral neuronal system, or the hematological system at the pharmacologically effective dose.
Co-reporter:Leli Zeng;Dr. Yu Chen;Huaiyi Huang;Jinquan Wang;Donglei Zhao; Liangnian Ji ;Dr. Hui Chao
Chemistry - A European Journal 2015 Volume 21( Issue 43) pp:15308-15319
Publication Date(Web):
DOI:10.1002/chem.201502154
Abstract
Hypoxia is the critical feature of the tumor microenvironment that is known to lead to resistance to many chemotherapeutic drugs. Six novel ruthenium(II) anthraquinone complexes were designed and synthesized; they exhibit similar or superior cytotoxicity compared to cisplatin in hypoxic HeLa, A549, and multidrug-resistant (A549R) tumor cell lines. Their anticancer activities are related to their lipophilicity and cellular uptake; therefore, these physicochemical properties of the complexes can be changed by modifying the ligands to obtain better anticancer candidates. Complex 1, the most potent member of the series, is highly active against hypoxic HeLa cancer cells (IC50=0.53 μM). This complex likely has 46-fold better activity than cisplatin (IC50=24.62 μM) in HeLa cells. This complex tends to accumulate in the mitochondria and the nucleus of hypoxic HeLa cells. Further mechanistic studies show that complex 1 induced cell apoptosis during hypoxia through multiple pathways, including those of DNA damage, mitochondrial dysfunction, and the inhibition of DNA replication and HIF-1α expression, making it an outstanding candidate for further in vivo studies.
Co-reporter:Dr. Li Xu;Xiang Chen;Jingheng Wu;Jinquan Wang; Liangnian Ji;Dr. Hui Chao
Chemistry - A European Journal 2015 Volume 21( Issue 10) pp:4008-4020
Publication Date(Web):
DOI:10.1002/chem.201405991
Abstract
A series of dinuclear ruthenium(II) complexes were synthesised, and the complexes were determined to be new highly selective compounds for binding to telomeric G-quadruplex DNA. The interactions of these complexes with telomeric G-quadruplex DNA were studied by using circular dichroism (CD) spectroscopy, fluorescence resonance energy transfer (FRET) melting assays, isothermal titration calorimetry (ITC) and molecular modelling. The results showed that the complexes 1, 2 and 4 induced and stabilised the formation of antiparallel G-quadruplexes of telomeric DNA in the absence of salt or in the presence of 100 mM K+-containing buffer. Furthermore, complexes 1 and 2 strongly bind to and effectively stabilise the telomeric G-quadruplex structure and have significant selectivity for G-quadruplex over duplex DNA. In comparison, complex 3 had a much lesser effect on the G-quadruplex, suggesting that possession of a suitably sized plane for good π–π stacking with the G-quadruplets is essential for the interaction of the dinuclear ruthenium(II) complexes with the G-quadruplex. Moreover, telomerase inhibition by the four complexes and their cellular effects were studied, and complex 1 was determined to be the most promising inhibitor of both telomerase and HeLa cell proliferation.
Co-reporter:Pingyu Zhang, Huaiyi Huang, Yu Chen, Jinquan Wang, Liangnian Ji, Hui Chao
Biomaterials 2015 53() pp: 522-531
Publication Date(Web):
DOI:10.1016/j.biomaterials.2015.02.126
Co-reporter:Xiang Chen, Lingli Sun, Yu Chen, Xiaolin Cheng, Weijun Wu, Liangnian Ji, Hui Chao
Biomaterials 2015 58() pp: 72-81
Publication Date(Web):
DOI:10.1016/j.biomaterials.2015.04.012
Co-reporter:Jiangping Liu, Yu Chen, Guanying Li, Pingyu Zhang, Chengzhi Jin, Leli Zeng, Liangnian Ji, Hui Chao
Biomaterials 2015 56() pp: 140-153
Publication Date(Web):
DOI:10.1016/j.biomaterials.2015.04.002
Co-reporter:Guanying Li, Qian Lin, Lingli Sun, Changsheng Feng, Pingyu Zhang, Bole Yu, Yu Chen, Ya Wen, Hui Wang, Liangnian Ji, Hui Chao
Biomaterials 2015 53() pp: 285-295
Publication Date(Web):
DOI:10.1016/j.biomaterials.2015.02.106
Co-reporter:Dr. Bole Yu;Cheng Ouyang;Kangqiang Qiu; Jing Zhao; Liangnian Ji; Hui Chao
Chemistry - A European Journal 2015 Volume 21( Issue 9) pp:3691-3700
Publication Date(Web):
DOI:10.1002/chem.201405151
Abstract
Fluorescence detection is the most effective tool for tracking gene delivery in living cells. To reduce photodamage and autofluorescence and to increase deep penetration into cells, choosing appropriate fluorophores that are capable of two-photon activation under irradiation in the NIR or IR regions is an effective approach. In this work, we have developed six tetranuclear ruthenium(II) complexes, GV1–6, and have studied their one- and two-photon luminescence properties. DNA interaction studies have demonstrated that GV2–6, bearing hydrophobic alkyl ether chains, show more efficient DNA condensing ability but lower DNA binding constants than GV1. However, the hydrophobic alkyl ether chains also enhance the DNA delivery ability of GV2–6 compared with that of GV1. More importantly, we have applied GV1–6 as non-viral gene vectors for tracking DNA delivery in living cells by one- and two-photon fluorescence microscopies. In two-photon microscopy, a high signal-to-noise contrast was achieved by irradiation with an 830 nm laser. This is the first example of the use of transition-metal complexes for two-photon luminescent tracking of the cellular pathways of gene delivery and as DNA carriers. Our work provides new insights into improving real-time tracking during gene delivery and transfection as well as important information for the design of multifunctional non-viral vectors.
Co-reporter:Huaiyi Huang;Pingyu Zhang;Hongmin Chen; Liangnian Ji ;Dr. Hui Chao
Chemistry - A European Journal 2015 Volume 21( Issue 2) pp:715-725
Publication Date(Web):
DOI:10.1002/chem.201404922
Abstract
The aim of this study was to illustrate the dramatically different anticancer activities between coordinatively saturated polypyridyl (1 a–4 a) and cyclometalated (1 b–4 b) ruthenium(II) complexes. The cyclometalated complexes 1 b–4 b function as DNA transcription inhibitors, exhibiting switch-on cytotoxicity against a 2D cancer cell monolayer, whereas the polypyridyl complexes 1 a–4 a are relatively inactive. Moreover, complexes 1 b–4 b exhibit excellent cytotoxicity against 3D multicellular tumor spheroids (MCTSs), which serve as an intermediate model between in vitro 2D cell monolayers and in vivo 3D solid tumors. The hydrophobicity, efficient cell uptake, and nucleus targeting ability, as well as the high DNA binding affinity of complexes 1 b–4 b, likely contribute to their enhanced anticancer activity. We surmise that cyclometalation could be a universal approach to significantly enhance the anticancer activity of substituted polypyridyl RuII complexes. We also suggest that 3D MCTSs may be a more practical platform for anticancer drug screening than 2D cancer monolayer approaches.
Co-reporter:Chengzhi Jin;Jiangping Liu;Dr. Yu Chen;Leli Zeng;Ruilin Guan;Cheng Ouyang; Liangnian Ji ;Dr. Hui Chao
Chemistry - A European Journal 2015 Volume 21( Issue 34) pp:
Publication Date(Web):
DOI:10.1002/chem.201583462
Co-reporter:Chengzhi Jin;Jiangping Liu;Dr. Yu Chen;Leli Zeng;Ruilin Guan;Cheng Ouyang; Liangnian Ji ;Dr. Hui Chao
Chemistry - A European Journal 2015 Volume 21( Issue 34) pp:12000-12010
Publication Date(Web):
DOI:10.1002/chem.201501882
Abstract
Five cyclometalated iridium(III) complexes with 2-phenylimidazo[4,5-f][1,10]phenanthroline derivatives (IrL1–IrL5) were synthesized and developed to image and track mitochondria in living cells under two-photon (750 nm) excitation, with two-photon absorption cross-sections of 48.8–65.5 GM at 750 nm. Confocal microscopy and inductive coupled plasma-mass spectrometry (ICP-MS) demonstrated that these complexes selectively accumulate in mitochondria within 5 min, without needing additional reagents for membrane permeabilization, or replacement of the culture medium. In addition, photobleaching experiments and luminescence measurements confirmed the photostability of these complexes under continuous laser irradiation and physiological pH resistance. Moreover, results using 3D multicellular spheroids demonstrate the proficiency of these two-photon luminescent complexes in deep penetration imaging. Two-photon excitation using such novel complexes of iridium(III) for exclusive visualization of mitochondria in living cells may substantially enhance practical applications of bioimaging and tracking.
Co-reporter:Guan-Ying Li, Rui-Lin Guan, Liang-Nian Ji, Hui Chao
Coordination Chemistry Reviews 2014 Volume 281() pp:100-113
Publication Date(Web):1 December 2014
DOI:10.1016/j.ccr.2014.09.005
•An overview of DNA condensation induced by metal complexes is provided.•DNA condensing mechanisms of metal complexes are summarised.•Some applications for these metal complexes in gene vectors are presented.•Some applications for these metal complexes in antitumor agents are presented.DNA is stored in a highly compact, condensed phase in viruses, bacteria and eukaryotes. Co(NH3)63+ is a well-known inorganic cation that can induce DNA condensation, and numerous Co(III), Co(II), Fe(II), Ca(II), Cu(II), Ni(II), Zn(II), Ru(II), Pt(II) and La(III) complexes have been evaluated as DNA condensing agents. In this review, we divided these metal complexes into four distinct classes based on the mechanism of DNA condensation: (i) complexes with high positive charges that condense DNA via electrostatic interaction, (ii) complexes with planar intercalative ligands that condense DNA via π–π interaction, (iii) complexes that bind with DNA through hydrogen bonds, and (iv) complexes that covalently bind to DNA. Some applications of these metal complex-based DNA condensing agents in gene vectors and antitumor agents are also presented.
Co-reporter:Guanying Li, Qian Lin, Liangnian Ji and Hui Chao
Journal of Materials Chemistry A 2014 vol. 2(Issue 45) pp:7918-7926
Publication Date(Web):18 Sep 2014
DOI:10.1039/C4TB01251H
Mitochondria are one of the major sources of cellular reactive oxygen species (ROS) and the hypochlorite ion (ClO−) is one kind of highly ROS involved in many important biological events. Herein, we present a novel class of mitochondrial ClO− probes. A series of iridium(III) complexes, Ir1–Ir4, were synthesized which exhibited a highly specific response of their phosphorescence signal toward ClO− over other ROS. By changing the cyclometalated ligands, the turn-on phosphorescent emission colours can be tuned from green to red. ICP-MS results and Mito-tracker co-staining experiments revealed that Ir1–Ir4 localized specifically in mitochondria. Finally, complexes Ir1–Ir4 were successfully applied in subcellular imaging for detection of endogenous ClO− in mitochondria.
Co-reporter:Wenchao Xu, Jiarui Zuo, Lili Wang, Liangnian Ji and Hui Chao
Chemical Communications 2014 vol. 50(Issue 17) pp:2123-2125
Publication Date(Web):02 Jan 2014
DOI:10.1039/C3CC48916G
A new series of dinuclear ruthenium(II) polypyridyl complexes, which possess larger π-conjugated systems, good water solubility and pH resistance, and high photostability, were developed to act as single and two-photon luminescence cellular imaging probes.
Co-reporter:Huaiyi Huang ; Pingyu Zhang ; Bole Yu ; Yu Chen ; Jinquan Wang ; Liangnian Ji
Journal of Medicinal Chemistry 2014 Volume 57(Issue 21) pp:8971-8983
Publication Date(Web):October 14, 2014
DOI:10.1021/jm501095r
Recently, coordinatively saturated and substitutionally inert Ru(II) complexes have been investigated as anticancer agents. Herein a cyclometalated Ru(II) complex, [Ru(bpy)(phpy)(dppz)]+, was found to be rapidly taken up by cancer cells, and nearly 90% of the complex accumulated in the nuclei of cancer cells after a 2 h incubation. The anticancer activity of this complex was screened against a panel of cancer cell lines. Remarkably, it exhibited IC50 values that were an order of magnitude lower than those of cisplatin. This complex also displayed potencies superior to those of cisplatin against 3D tumor spheroids. Further studies revealed that the high DNA binding affinity of [Ru(bpy)(phpy)(dppz)]+ resulted in effective disruption of the binding of transcription factor NF-κB to DNA sequences, thereby inhibiting cellular transcription and leading to irreversible cancer cell apoptosis. Our work provides new insights into understanding the biological interactions and anticancer molecular mechanisms of DNA-specific Ru(II) polypyridyl complexes.
Co-reporter:Guoliang Liao, Xiang Chen, Jingheng Wu, Chen Qian, Hanqiang Wang, Liangnian Ji and Hui Chao
Dalton Transactions 2014 vol. 43(Issue 21) pp:7811-7819
Publication Date(Web):10 Mar 2014
DOI:10.1039/C3DT53547A
Two novel ruthenium polypyridyl complexes, Ru[(bpy)2(pedppz)]2+ (1) and Ru[(bpy)2(pemitatp)]2+ (2) (bpy = 2′2-bipyridine, pdeppz = 10-(2-(piperidin-1-yl)ethoxy)dipyrido[3,2-a:2′,3′-c]phenazine, pemitatp = 5-methoxy-1-(2-(piperidin-1-yl)ethyl)-isatino[1,2-b]-1,4,8,9-tetraazatriphenylene), bearing large planar π-delocalized aromatic systems with flexible chains have been synthesised and characterised. As expected, these complexes show inhibition towards telomerase by inducing and stabilising the G-quadruplex structure.
Co-reporter:Hui Chao
Journal of Inorganic Biochemistry 2014 Volume 138() pp:144
Publication Date(Web):September 2014
DOI:10.1016/j.jinorgbio.2014.05.004
Co-reporter:Jie-Wen Liang, Yi Wang, Ke-Jie Du, Guan-Ying Li, Rui-Lin Guan, Liang-Nian Ji, Hui Chao
Journal of Inorganic Biochemistry 2014 Volume 141() pp:17-27
Publication Date(Web):December 2014
DOI:10.1016/j.jinorgbio.2014.08.006
Three novel copper(II) complexes CuL1Cl2 (1) (L1 = 4′-(3-methoxyphenyl)-2,2′:6′- 2″-terpyridine), CuL2Cl2 (2) (L2 = 4′-(4-methoxyphenyl)-2,2′:6′-2″-terpyridine) and CuL3Cl2 (3) (L3 = 4′-(3,5-dimethoxyphenyl)-2,2′:6′-2″-terpyridine) have been synthesized and characterized. Absorption spectral titration experiments, ethidium bromide displacement assays, and cyclic voltammetric experiments were carried out and the results suggested that these complexes bound to DNA through an intercalative mode. Moreover, these complexes were found to cleave pBR322 DNA efficiently in the presence of glutathione (GSH), and exhibited good anticancer activity against HeLa, Hep-G2 and BEL-7402 cell lines. Nuclear chromatin cleavage was also observed by acridine orange/ethidium bromide (AO/EB) staining assays and comet assays. These results demonstrated that these three Cu(II) complexes caused DNA damage and induced the apoptosis of HeLa cells. Mechanistic investigations revealed the participation of reactive oxygen species which can be trapped by reactive oxygen species (ROS) radical scavengers and ROS sensors.Three copper(II) complexes cleaved DNA efficiently in the presence of GSH and bounded to DNA through intercalation mode. They can also induce the apoptosis of HeLa cells with the participation of reactive oxygen species.
Co-reporter:Yu-Chuan Wang, Chen Qian, Zai-Li Peng, Xiao-Juan Hou, Li-Li Wang, Hui Chao, Liang-Nian Ji
Journal of Inorganic Biochemistry 2014 130() pp: 15-27
Publication Date(Web):
DOI:10.1016/j.jinorgbio.2013.09.015
Co-reporter:Jin-Quan Wang;Ping-Yu Zhang;Chen Qian
JBIC Journal of Biological Inorganic Chemistry 2014 Volume 19( Issue 3) pp:335-348
Publication Date(Web):2014 March
DOI:10.1007/s00775-013-1069-2
A series of novel chiral ruthenium(II) polypyridyl complexes (Δ-Ru1, Λ-Ru1, Δ-Ru2, Λ-Ru2, Δ-Ru3, Λ-Ru3) were synthesized and evaluated to determine their antiproliferative activities. Colocalization, inductively coupled plasma mass spectrometry, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay studies showed that these ruthenium(II) complexes accumulated preferentially in the mitochondria and exhibited cytotoxicity against various cancer cells in vitro. The complex Δ-Ru1 is of particular interest because it was found to have half-maximal inhibitory concentrations comparable to those of cisplatin and better activity than cisplatin against a cisplatin-resistant cell line, A549-CP/R. Δ-Ru1 induced alterations in the mitochondrial membrane potential and triggered intrinsic mitochondria-mediated apoptosis in HeLa cells, which involved the regulation of Bcl-2 family members and the activation of caspases. Taken together, these data suggest that Δ-Ru1 may be a novel mitochondria-targeting anticancer agent.
Co-reporter:Pingyu Zhang, Jinquan Wang, Huaiyi Huang, Hongming Chen, Ruilin Guan, Yu Chen, Liangnian Ji, Hui Chao
Biomaterials 2014 35(32) pp: 9003-9011
Publication Date(Web):
DOI:10.1016/j.biomaterials.2014.07.021
Co-reporter:Yu Chen, Liping Qiao, Liangnian Ji, Hui Chao
Biomaterials 2014 35(1) pp: 2-13
Publication Date(Web):
DOI:10.1016/j.biomaterials.2013.09.051
Co-reporter:Guanying Li, Yu Chen, Jinquan Wang, Qian Lin, Jing Zhao, Liangnian Ji and Hui Chao
Chemical Science 2013 vol. 4(Issue 12) pp:4426-4433
Publication Date(Web):02 Sep 2013
DOI:10.1039/C3SC52301B
A non-emissive dinuclear iridium(III) complex (SP-2) linked via an azo group has been adopted as a phosphorescent probe for sulphite and bisulphite. SP-2 exhibits a turn-on phosphorescent response toward sulphite and bisulphite with high selectivity and sensitivity. Furthermore, this probe can be used for living cell imaging to identify the presence of both external sulphite/bisulphite supplemented into cell cultures and endogenous sulphite/bisulphite biosynthesised in living cells.
Co-reporter:Yu Chen, Liping Qiao, Bole Yu, Guanying Li, Chunyuan Liu, Liangnian Ji and Hui Chao
Chemical Communications 2013 vol. 49(Issue 94) pp:11095-11097
Publication Date(Web):08 Oct 2013
DOI:10.1039/C3CC46957C
An AIPE-active iridium(III) complex was found to possess high specificity for mitochondria, superior photostability, low cytotoxicity, and high resistance to the loss of mitochondrial membrane potential. Thus, this complex can be used for mitochondrial imaging and tracking in living cells.
Co-reporter:Bole Yu, Yu Chen, Cheng Ouyang, Huaiyi Huang, Liangnian Ji and Hui Chao
Chemical Communications 2013 vol. 49(Issue 8) pp:810-812
Publication Date(Web):06 Dec 2012
DOI:10.1039/C2CC37896E
A luminescent tetranuclear ruthenium(II) complex was developed to act as a DNA carrier and at the same time offer luminescent imaging to follow the DNA intracellular trafficking with time.
Co-reporter:Guanying Li, Yu Chen, Jingheng Wu, Liangnian Ji and Hui Chao
Chemical Communications 2013 vol. 49(Issue 20) pp:2040-2042
Publication Date(Web):25 Jan 2013
DOI:10.1039/C3CC38687B
Based on an azo–thiol redox reaction, a non-emissive dinuclear iridium(III) complex showed off–on phosphorescent response toward thiols and was developed to image thiol levels in living cells.
Co-reporter:Xiang Chen, Jing-Heng Wu, Ying-Wei Lai, Rong Zhao, Hui Chao and Liang-Nian Ji
Dalton Transactions 2013 vol. 42(Issue 13) pp:4386-4397
Publication Date(Web):10 Jan 2013
DOI:10.1039/C3DT32921F
Two ruthenium(II) polypyridyl complexes, [Ru(bpy)2(ptpn)]2+ (1) (bpy = 2,2′-bipyridine, ptpn = 3-(1,10-phenanthroline-2-yl)-as-triazino[5,6-f]1,10-phenanthroline) and [Ru(phen)2(ptpn)]2+ (2) (phen = 1,10-phenanthroline), were synthesized and characterized. Crystal structure analysis shows that complex 1 has a large planar aromatic area and possesses the potential to fit the geometric structure of G-quadruplex. The interaction of the G-quadruplex DNA with Ru(II) complexes was explored by means of circular dichroism (CD), fluorescence resonance energy transfer (FRET) melting assay, competitive FRET assay and polymerase chain reaction (PCR) stop assay. The results indicated that complexes 1 and 2 both have the ability to promote the formation and stabilization of the human telomeric d[(TTAGGG)n] (HTG22) quadruplex and exhibit high G-quadruplex DNA selectivity over duplex DNA. The telomere repeat amplification protocol (TRAP) assay and long-term proliferation experiments further demonstrate that the Ru(II) complexes are potent telomerase inhibitors and HeLa cell proliferation inhibitors.
Co-reporter:Pingyu Zhang, Jinquan Wang, Huaiyi Huang, Liping Qiao, Liangnian Ji and Hui Chao
Dalton Transactions 2013 vol. 42(Issue 24) pp:8907-8917
Publication Date(Web):18 Apr 2013
DOI:10.1039/C3DT50472G
A series of novel chiral ruthenium(II) complexes with phenolic hydroxyl groups were synthesized and characterized. These ruthenium(II) complexes exhibited strong dual inhibition of topoisomerases I and IIα, with approximate IC50 values of 3–15 mM, which were more efficient than the widely clinically used single TopoI poison camptothecin (CPT) or TopoIIα poison etoposide (VP-16). Δ-1 and Λ-1 with more hydroxyls were observed to be more potent inhibitors. To further evaluate the mechanism of the complexes at a cellular level, these complexes were investigated for their effect on cell proliferation, cell cycle progression and induction of apoptosis. The results indicated that ruthenium(II) complexes permeated the nuclei in cancer cells and inhibited the activities of nuclear enzymes topoisomerases I and IIα, then triggered DNA damage and induced apoptosis in the cancer cells. The simultaneous inhibition of TopoI and TopoIIα induced the death of cancer cells, which may be a promising and effective strategy for cancer therapy.
Co-reporter:Chen Qian, Jin-Quan Wang, Cui-Lan Song, Li-Li Wang, Liang-Nian Ji and Hui Chao
Metallomics 2013 vol. 5(Issue 7) pp:844-854
Publication Date(Web):20 Feb 2013
DOI:10.1039/C3MT20270D
Four ruthenium(II) asymmetric complexes, [Ru(bpy)2(PAIDH)]2+ (bpy = 2,2′-bipyridine, PAIDH = 2-pyridyl-1H-anthra[1,2-d]imidazole-6,11-dione, 1), [Ru(phen)2(PAIDH)]2+ (phen = 1,10-phenanthroline, 2), [Ru(dmp)2(PAIDH)]2+ (dmp = 4,7-dimethyl-1,10-phenanthroline, 3) and [Ru(dip)2(PAIDH)]2+ (dip = 4,7-diphenyl-1,10-phenanthroline, 4), have been synthesized and characterized. These complexes displayed potent anti-proliferation activity against various cancer cell lines and had high selectivity between tumor cells and normal cells. HeLa cells exhibited the highest sensitivity to complex 4, accounting for the greatest cellular uptake. Complex 4 was shown to accumulate preferentially in the mitochondria of HeLa cells and induced apoptosis via the mitochondrial pathway, which involved ROS generation, mitochondrial membrane potential depolarisation, and Bcl-2 and caspase family members activation. These results demonstrated that complex 4 induced cancer cell apoptosis by acting on mitochondrial pathways.
Co-reporter:Lüying Li, Kejie Du, Yi Wang, Haina Jia, Xiaojuan Hou, Hui Chao and Liangnian Ji
Dalton Transactions 2013 vol. 42(Issue 32) pp:11576-11588
Publication Date(Web):04 Jun 2013
DOI:10.1039/C3DT50395J
Three mononuclear copper complexes [Cu(PDTP)Cl2] (PDTP = 4-phenyl-2,6-di(thiazole-2-yl)pyridine, CuPDTP), [Cu(ADTP)Cl2] (ADTP = 4-(anthracen-9-yl)-2,6-di(thiazole-2-yl)pyridine, CuADTP) and [Cu(BFDTP)Cl2] (BFDTP = 4-(benzofuran-2-yl)-2,6-di(thiazole-2-yl)pyridine, CuBFDTP) were synthesized and characterized. The X-ray single crystallography results indicated that the Cu(II) ions showed slightly distorted square pyramid coordination environments, and the ligands deviated from ideal planarity in all three compounds. Based on the DNA binding studies, it was demonstrated that these three complexes exhibited weak DNA binding strengths, which were most likely groove binding modes. CuPDTP, CuADTP and CuBFDTP induced efficient DNA cleavage in the dark without the addition of external catalysts (oxidant or reductant). In contrast, in the presence of reducing or oxidizing agents, the nuclease activities increased more than 10-fold. Mechanistic investigations revealed the participation of reactive oxygen species, which can be trapped by ROS radical scavengers and ROS sensors. In the same experimental conditions, the free ligands and CuCl2 did not display any DNA cleaving activity. This result indicates that the complexes, rather than their components, play a significant role in the nuclease reaction process and that DNA cleavage may be initiated in an oxidative pattern. The proposed mechanism was attributed to the in situ activation of molecular oxygen by the oxidation of the copper complexes. In the MTT cytotoxicity studies, the three Cu(II) complexes exhibited an antitumor activity against the HeLa, BEL-7402 and HepG2 tumor cell lines. The HeLa cells treated with Cu(II) complexes demonstrated marked changes in their nuclear morphology, which were detected by Hoechst 33258 nuclear staining and acridine orange/ethidium bromide (AO/EB) staining assays. Nuclear chromatin cleavage also was observed from alkaline single-cell gel electrophoresis (comet assay).
Co-reporter:Guan-Ying Li, Jiang-Ping Liu, Huai-Yi Huang, Ya Wen, Hui Chao, Liang-Nian Ji
Journal of Inorganic Biochemistry 2013 Volume 121() pp:108-113
Publication Date(Web):April 2013
DOI:10.1016/j.jinorgbio.2012.12.019
A dinuclear ruthenium(II) complex linked via a reducible azo group [Ru(bpy)2(azobpy)Ru(bpy)2]Cl4 (Ru2azo, bpy = 2,2′-bipyridine, azobpy = 4,4″-azobis (2,2′-bipyridine)) was adopted as a probe for thiols. Results showed that Ru2azo could selectively and effectively react with biological thiols (such as cysteine, homocysteine and glutathione) with a 10− 7 M detection limit. After it reacted with thiols, the original gray color of Ru2azo solution immediately turned yellow and the luminescence significantly enhanced, showing “naked-eye” colorimetric and “off–on” luminescent dual-signaling response for thiols. Mechanism studies demonstrated that Ru2azo reacted with thiols undergoing a two-electron transfer process, forming the azo2 − anion product.A dinuclear ruthenium(II) complex [Ru(bpy)2(azobpy)Ru(bpy)2]Cl4 (Ru2azo) could selectively and effectively detect biological thiols with a 10− 7 mol/L detection limit. After it reacted with thiols, the original gray color of Ru2azo solution immediately turned yellow and the luminescence significantly enhanced, exhibiting “naked-eye” colorimetric and “off-on” luminescent dual-signaling response for thiols.Highlights► A ruthenium(II)-azo complex-based thiol probe was developed. ► Ru(II) complex can selectively react with thiols with a 10− 7 M detection limit. ► Ru(II) complexes exhibited “naked-eye” colorimetric response for thiols. ► Ru(II) complexes exhibited “off–on” luminescent switch for thiols.
Co-reporter:Guan-Ying Li, Ke-Jie Du, Jin-Quan Wang, Jie-Wen Liang, Jun-Feng Kou, Xiao-Juan Hou, Liang-Nian Ji, Hui Chao
Journal of Inorganic Biochemistry 2013 Volume 119() pp:43-53
Publication Date(Web):February 2013
DOI:10.1016/j.jinorgbio.2012.09.019
Three new tridentate copper(II) complexes [Cu(dthp)Cl2] (1) (dthp = 2,6-di(thiazol-2-yl)pyridine), [Cu(dmtp)Cl2] (2) (dmtp = 2,6-di(5-methyl-4H-1,2,4-triazol-3-yl)pyridine) and [Cu(dtp)Cl2] (3) (dtp = 2,6-di(4H-1,2,4-triazol-3-yl)pyridine) have been synthesized and characterized. Crystal structure of complex 1 shows that the complex existed as distorted square pyramid with five co-ordination sites occupied by the tridentate ligand and the two chlorine anions. Ethidium bromide displacement assay, viscosity measurements, circular dichroism studies and cyclic voltammetric experiments suggested that these complexes bound to DNA via an intercalative mode. Three Cu(II) complexes were found to efficiently cleave DNA in the presence of sodium ascorbate, and singlet oxygen (1O2) and hydrogen peroxide were proved to contribute to the DNA cleavage process. They exhibited anticancer activity against HeLa, Hep-G2 and BEL-7402 cell lines. Nuclear chromatin cleavage has also been observed with AO/EB staining assay and the alkaline single-cell gel electrophoresis (comet assay). The results demonstrated that three Cu(II) complexes cause DNA damage that can induce the apoptosis of BEL-7402 cells.Three copper(II) complexes [Cu(dthp)Cl2] (dthp = 2,6-di(thiazol-2-yl)pyridine), [Cu(dmtp)Cl2] (dmtp = 2,6-di(5-methyl-4H-1,2,4-triazol-3-yl)pyridine) and [Cu(dtp)Cl2] (dtp = 2,6-di(4H-1,2,4-triazol-3-yl)pyridine) were found to efficiently cleave DNA in the presence of sodium ascorbate. They can induce the apoptosis of BEL-7402 cells.Figure optionsDownload full-size imageDownload as PowerPoint slideHighlights► Synthesis of complexes [Cu(dthp)Cl2], [Cu(dmtp)Cl2] and [Cu(dtp)Cl2]. ► Three Cu(II) complexes bind to DNA in an intercalative mode. ► Cu(II) complexes efficiently cleave DNA in the presence of sodium ascorbate. ► Cu(II) complexes can induce the apoptosis of BEL-7402 cells.
Co-reporter:Pingyu Zhang;Dr. Lingmin Pei;Dr. Yu Chen;Wenchao Xu;Qitian Lin;Jinquan Wang;Jingheng Wu;Dr. Yong Shen; Liangnian Ji ; Hui Chao
Chemistry - A European Journal 2013 Volume 19( Issue 46) pp:15494-15503
Publication Date(Web):
DOI:10.1002/chem.201302919
Abstract
A new dinuclear RuII polypyridyl complex, [(bpy)2Ru(H2bpip)Ru(bpy)2]4+ (RuH2bpip, bpy=2,2-bipyridine, H2bpip=2,6-pyridyl(imidazo[4,5-f][1,10]phenanthroline), was developed to act as a one- and two-photon luminescent probe for biological Cu2+ detection. This RuII complex shows a significant two-photon absorption cross section (400 GM) and displays a remarkable one- and two-photon luminescence switch in the presence of Cu2+ ions. Importantly, RuH2bpip can selectively recognise Cu2+ in aqueous media in the presence of other abundant cellular cations (such as Na+, K+, Mg2+, and Ca2+), trace metal ions in organisms (such as Zn2+, Ag+, Fe3+, Fe2+, Ni2+, Mn2+, and Co2+), prevalent toxic metal ions in the environment (such as Cd2+, Hg2+, and Cr3+), and amino acids, with high sensitivity (detection limit≤3.33×10−8 M) and a rapid response time (≤15 s). The biological applications of RuH2bpip were also evaluated and it was found to exhibit low cytotoxicity, good water solubility, and membrane permeability; RuH2bpip was, therefore, employed as a sensing probe for the detection of Cu2+ in living cells and zebrafish.
Co-reporter:Guo-Liang Liao, Xiang Chen, Liang-Nian Ji and Hui Chao
Chemical Communications 2012 vol. 48(Issue 87) pp:10781-10783
Publication Date(Web):13 Sep 2012
DOI:10.1039/C2CC36039J
A novel ruthenium(II) complex with specific luminescent selectivity towards hybrid G-quadruplex DNA was developed that can easily be distinguished by the naked eye without further treatment.
Co-reporter:Lü-Ying Li, Hai-Na Jia, Hui-Juan Yu, Ke-Jie Du, Qi-Tian Lin, Kang-Qiang Qiu, Hui Chao, Liang-Nian Ji
Journal of Inorganic Biochemistry 2012 Volume 113() pp:31-39
Publication Date(Web):August 2012
DOI:10.1016/j.jinorgbio.2012.03.008
Two ruthenium(II) polypyridyl complexes [Ru(tpy)(ptn)]2 + (1) and Ru(dmtpy)(ptn)]2 + (2) (ptn = 3-(1,10-phenanthrolin-2-yl)-as-triazino[5,6-f]naphthalene, tpy = 2,2’:6’,2”-terpyridine, dmtpy = 5,5’-dimethyl-2,2’:6’,2”-terpyridine) have been synthesized and characterized by elemental analysis, 1 H NMR, mass spectrometry and crystal structure analysis. Spectroscopic studies together with isothermal titration calorimetry (ITC) and viscosity measurements prove that two complexes bind to DNA in an intercalative mode. ITC experiments show that the binding mode for complex 2 is entropically driven, while an entropy-driven initial binding of complex 1 is followed by an entropically and enthalpically favorable process. This difference may be attributed to the ancillary ligand effects on the DNA binding of Ru(II) complexes. Circular dichroism titrations of calf thymus DNA (CT-DNA) with Ru(II) complexes show that complexes 1 and 2 induce B to Z conformational transition of calf thymus DNA at low ionic strength (0.05 M NaCl). The induced Z-DNA conformation can revert to B form when Ru(II) complexes are displaced by ethidium bromide or at high ionic strengths ([NaCl] = 0.4 M), but keeps intact with temperature ranged from 25 to 90 °C. The unique structure and characteristics of Ru(II) complexes designed in this investigation will be useful for the study of Z-DNA.Two DNA-intercalating ruthenium(II) complexes [Ru(tpy)(ptn)]2 + (1) and Ru(dmtpy)(ptn)]2 + (2) exhibit a remarkable ability to induce the B-Z transition of CT-DNA at low ionic strength. The induced Z-DNA conformation keeps intact with temperature ranged from 25 to 90 °C.Highlights► Synthesis of two new Ru(II) complexes [Ru(tpy)(ptn)]2+ and Ru(dmtpy)(ptn)]2+. ► Two Ru(II) complexes bind to DNA in an intercalative mode. ► Two complexes induce B-Z transition in CT-DNA at low ionic strength (0.05 M NaCl). ► The induced Z-DNA conformation keeps intact from 25 to 90 °C.
Co-reporter:Qi-Tian Lin, Ling-Min Pei, Wen-Chao Xu, Hui Chao, Liang-Nian Ji
Inorganic Chemistry Communications 2012 Volume 16() pp:104-106
Publication Date(Web):February 2012
DOI:10.1016/j.inoche.2011.11.036
A novel ruthenium(II) polypyridyl complex [Ru(bpy)2(pipdpa)]2+ (1) with DPA (dipicolylamine) moiety as receptor (bpy = 2,2′-bipyridine, pipdpa=2-(4′-N,N-bis(pyridin-2-ylmethyl)aniline)imidazo[4,5-f][1,10]phenanthroline) was developed as a luminescent chemosensor with high selectivity and antidisturbance toward Cu2+ over other tested metal ions (Ca2+, Cd2+, Co2+, Hg2+, K+, Mg2+, Mn2+, Na+, Ni2+, Pb2+, Zn2+, Fe2+, Fe3+). Remarkably, complex 1 could be applied to detect Cu2+ with a wide pH range 3–10 in aqueous solution.A novel ruthenium(II) polypyridyl complex [Ru(bpy)2(pipdpa)]2+ (1) was developed as a highly sensitive and selective luminescent chemosensor for Cu2+ in aqueous solution.Highlights► [Ru(bpy)2(pipdpa)]2+ (1) was developed as a luminescent chemosensor for Cu2+. ► Complex 1 showed high sensitivity and selectivity toward Cu2+. ► Complex 1 displayed well solubility in aqueous solution. ► Complex 1 could be applied to detect Cu2+ with a wide pH range 3–10.
Co-reporter:Ling-Min Pei, Qi-Tian Lin, Hui Chao, Liang-Nian Ji
Inorganic Chemistry Communications 2012 Volume 22() pp:90-92
Publication Date(Web):August 2012
DOI:10.1016/j.inoche.2012.05.030
A new ruthenium(II) polypyridyl complex [Ru(tpy)(H4bbdip)]2+ (1) (tpy = terpyridine, H4bbdip = 2,6-bis(benzo[1,2-d:4,5-d′]diimidazole-2′-yl)pyridine) was developed as an optical sensor with selective colorimetric responses toward F− and OAc− over other tested anions (Cl−, Br−, I−, NO3−, HSO4−, H2PO4−).A new ruthenium(II) polypyridyl complex [Ru(tpy)(H4bbdip)]2+ (1) was developed as an optical sensor with selective colorimetric responses toward F− and OAc− over other tested anions (Cl−, Br−, I−, NO3−, HSO4−, H2PO4−).Highlights► Synthesis of a new ruthenium(II) polypyridyl complexes [Ru(tpy)(H4bbdip)]2+. ► [Ru(tpy)(H4bbdip)]2+ was developed as a colorimetric sensor for F− and OAc−. ► UV-vis and 1H NMR titrations were performed to investigate the interaction between anions and Ru(II) complex.
Co-reporter:Jun-Feng Kou;Chen Qian;Jin-Quan Wang
JBIC Journal of Biological Inorganic Chemistry 2012 Volume 17( Issue 1) pp:81-96
Publication Date(Web):2012 January
DOI:10.1007/s00775-011-0831-6
DNA topoisomerases (I and II) have been one of the excellent targets in anticancer drug development. Here two chiral ruthenium(II) anthraquinone complexes, Δ- and Λ-[Ru(bpy)2(ipad)]2+, where bpy is 2,2′-bipyridine and ipad is 2-(anthracene-9,10-dione-2-yl)imidazo[4,5-f][1,10]phenanthroline, were synthesized and characterized. As expected, both of the Ru(II) complexes intercalate into DNA base pairs and possess an obviously greater affinity with DNA. Topoisomerase inhibition and DNA strand passage assay confirmed that the two complexes are efficient dual inhibitors of topoisomerases I and II by interference with the DNA religation. In MTT cytotoxicity studies, two Ru(II) complexes exhibited antitumor activity against HeLa, MCF-7, HepG2 and BEL-7402 tumor cell lines. Flow cytometry analysis shows an increase in the percentage of cells with apoptotic morphological features in the sub-G1 phase for Ru(II) complexes. Nuclear chromatin cleavage has also been observed from AO/EB staining assay and alkaline single-cell gel electrophoresis (comet assay). The results demonstrated that Δ- and Λ-[Ru(bpy)2(ipad)]2+ act as dual inhibitors of topoisomerases I and II, and cause DNA damage that can lead to cell cycle arrest and/or cell death by apoptosis.
Co-reporter:Ke-Jie Du, Jin-Quan Wang, Jun-Feng Kou, Guan-Ying Li, Li-Li Wang, Hui Chao, Liang-Nian Ji
European Journal of Medicinal Chemistry 2011 Volume 46(Issue 4) pp:1056-1065
Publication Date(Web):April 2011
DOI:10.1016/j.ejmech.2011.01.019
Two ruthenium(II) complexes [Ru(bpy)2(bfipH)]2+ (1) and [Ru(phen)2(bfipH)]2+ (2) have been synthesized and characterized. The DNA-binding behaviors of complexes were studied by using spectroscopic and viscosity measurements. Results suggested that the two complexes bind to DNA in an intercalative mode. Complexes 1 and 2 can efficiently photocleave pBR322 DNA in vitro under irradiation, singlet oxygen (1O2) was proved to contribute to the DNA photocleavage process. Topoisomerase inhibition and DNA strand passage assay confirmed that two Ru(II) complexes acted as efficient dual inhibitors of topoisomerases I and II. In MTT cytotoxicity studies, two Ru(II) complexes exhibited antitumor activity against BEL-7402, HeLa, MCF-7 tumor cells. The AO/EB staining assay indicated that Ru(II) complexes could induce the apoptosis of HeLa cells.Two ruthenium complexes [Ru(bpy)2(bfipH)]2+ (1) and [Ru(phen)2(bfipH)]2+ (2) were found to act as efficient dual inhibitors of topoisomerases I and II. Research highlights► Two novel DNA-intercalating ruthenium(II) complexes [Ru(bpy)2(bfipH)]2+ (1) and [Ru(phen)2(bfipH)]2+ (2) were confirmed to act as efficient dual inhibitors of topoisomerases I and II. ► Both complexes exhibited antitumor activity against BEL-7402, HeLa, MCF-7 tumor cells. ► The AO/EB staining assay testified that Ru(II) complexes induce the apoptosis of HeLa cells.
Co-reporter:Li Xu, Guo-Liang Liao, Xiang Chen, Cun-Yuan Zhao, Hui Chao, Liang-Nian Ji
Inorganic Chemistry Communications 2010 Volume 13(Issue 9) pp:1050-1053
Publication Date(Web):September 2010
DOI:10.1016/j.inoche.2010.06.008
Two new trinuclear ruthenium(II) complexes [(bpy)6Ru3(tpbip)]6+ (1) and [(bpy)6Ru3(tptaip)]6+ (2) (bpy = 2,2′-bipyridine, tpbip = 1,3,5-tris(1,10-phenanthroline-[5,6-d]imidazol-2-yl)-benzene, tptaip = 2,4,6-tri(1,10-phenanthroline-[5,6-d]imidazol-2-yl)-1,3,5-triazine) have been synthesized and characterized. The interaction of human telomeric DNA with Ru(II) complexes was explored by means of CD spectroscopy, fluorescence titration, ITC and FRET melting. Results indicated that two complexes not only induce a remarkable conformational change of human telomeric DNA, but also have the ability to stabilize the G-quadruplex.Two trinuclear ruthenium(II) complexes [(bpy)6Ru3(tpbip)]6+ (1) and [(bpy)6Ru3(tptaip)]6+ (2) not only induce a remarkable conformational change of human telomeric DNA, but also have the ability to stabilize the G-quadruplex.
Co-reporter:Yu Chen, Xu Zhou, Xu-Hui Wei, Bo-Le Yu, Hui Chao, Liang-Nian Ji
Inorganic Chemistry Communications 2010 Volume 13(Issue 9) pp:1018-1020
Publication Date(Web):September 2010
DOI:10.1016/j.inoche.2010.05.019
Three new ruthenium(II) polypyridyl complexes with highly π-deficient ligands, [Ru(bpy)2(btm)]2+ (1), [Ru(bpy)2(btb)]2+ (2) and [Ru(bpy)2(btp)]2+ (3) (bpy = 2,2′-bipyridine, btm = 3,3′-bis(5,6-dimethyl-1,2,4-triazine, btb = 3,3′-bis(5,6-diphenyl-1,2,4-triazine, btp = 3,3′-bis(phenanthro[9,10-e][1,2,4]triazine) were synthesized and characterized. Electrochemical data together with molecular calculations show that the first redox process in these complexes is not bpy based. Complexes 1, 2 and 3 display luminescence in ethanol/methanol (4/1, V/V) at 80 K, and all three complexes exhibit a temperature switch from single (150 K) to dual (80 K) emission behavior.Three ruthenium(II) complex [Ru(bpy)2(btm)]2+ (1), [Ru(bpy)2(btb)]2+ (2) and [Ru(bpy)2(btp)]2+ and (3) were found to exhibit a temperature switch from single (150 K) to dual (80 K) emission behavior.
Co-reporter:Yu Chen, Wen-Chao Xu, Jun-Feng Kou, Bo-Le Yu, Xu-Hui Wei, Hui Chao, Liang-Nian Ji
Inorganic Chemistry Communications 2010 Volume 13(Issue 10) pp:1140-1143
Publication Date(Web):October 2010
DOI:10.1016/j.inoche.2010.06.029
A new ruthenium(II) complex [Ru(bpy)2(pzta)]2+ (1) [bpy = 2,2′-bipyridine; pzta = 6-(pyrzin2-yl)-1,3,5-triazine-2,4-diamine] has been synthesized and characterized. This complex exhibits a typical aggregation-induced emission (AIE) behavior that it is faint in solution but intense in the aggregate state.A new ruthenium(II) complex [Ru(bpy)2(pzta)]2+ was found to exhibit aggregation-induced emission (AIE) behavior.
Co-reporter:Bin Sun, Yu-Chuan Wang, Chen Qian, Jun Chu, Si-Min Liang, Hui Chao, Liang-Nian Ji
Journal of Molecular Structure 2010 Volume 963(2–3) pp:153-159
Publication Date(Web):29 January 2010
DOI:10.1016/j.molstruc.2009.10.028
Two new chiral ruthenium(II) complexes Δ-[Ru(bpy)2(nfip)]2+ and Λ-[Ru(bpy)2(nfip)]2+ (in which bpy = 2,2′-bipyridine, nfip = 2-(5-nitrofuran-2-yl)-1H-imidazo[4,5-f][1,10]phenanthroline) have been synthesized and characterized by elemental analysis, ES-MS, 1H NMR and CD spectra. The binding properties of two chiral complexes with calf thymus DNA have been investigated by different spectrophotometric methods and viscosity measurements. A subtle but detectable difference was observed in the interaction of both enantiomer with CT-DNA, Δ enantiomer binds with DNA more strongly than Λ enantiomer. Viscosity experiments provided evidence that both Δ-[Ru(bpy)2(nfip)]2+ and Λ-[Ru(bpy)2(nfip)]2+ bound to DNA by intercalation.
Co-reporter:Yu Chen;WenChao Xu;JunFeng Kou;XuHui Wei;BoLe Yu
Science China Chemistry 2010 Volume 53( Issue 10) pp:2099-2105
Publication Date(Web):2010 October
DOI:10.1007/s11426-010-4099-4
Three Ru(II) complexes [Ru(bpy)2(1-IQTNH)](ClO4)2 (1), [Ru(bpy)2(2-QTNH)](ClO4)2 (2) and [Ru(bpy)2(3-IQTNH)](ClO4)2 (3) (bpy = 2,2′-bipyridine, 1-IQTNH = 6-(isoquinolin-1-yl)-1,3,5-triazine- 2,4-diamine, 2-QTNH = 6-(quinolin-2-yl)-1,3,5-triazine- 2,4-diamine, 3-IQTNH = 6-(isoquinolin-3-yl)-1,3,5-triazine-2,4-diamine) have been synthesized and characterized by elemental analysis, 1H NMR spectroscopy, electrospray ionization mass spectrometry and X-ray crystallography. The electrochemical and spectroscopic properties of the complexes differ from those of [Ru(bpy)3]2+ owing to the structural differences between the ligands and their complexes.
Co-reporter:Feng Gao, Xing Chen, Jin-Quan Wang, Yu Chen, Hui Chao and Liang-Nian Ji
Inorganic Chemistry 2009 Volume 48(Issue 13) pp:5599-5601
Publication Date(Web):June 8, 2009
DOI:10.1021/ic900902f
Three Ru(II) polypyridyl complexes with potential high DNA-binding ability have been designed and synthesized by extending the conjugated plane of the intercalative ligand and introducing electropositive pendants to the ancillary ligand. Spectral titration, DNA thermal denaturation, viscosity experiments, and quantum chemistry calculations were performed, and the complexes were found to intercalate into DNA base pairs with very high affinity even at high salt concentrations. Benefiting from their high DNA-binding ability, the complexes can effectively inhibit the DNA transcription activity by blocking the binding of T7 RNA polymerase to the template DNA. As efficient transcription inhibitors, the complexes demonstrated high in vitro antitumor activity against four selected tumor cell lines.
Co-reporter:Bin Sun, Jing-Xin Guan, Li Xu, Bo-Le Yu, Long Jiang, Jun-Feng Kou, Li Wang, Xi-Dong Ding, Hui Chao and Liang-Nian Ji
Inorganic Chemistry 2009 Volume 48(Issue 11) pp:4637-4639
Publication Date(Web):April 10, 2009
DOI:10.1021/ic900102r
Two novel DNA-intercalating ruthenium(II) complexes, [Ru(bpy)2(PIPSH)]2+ and [Ru(bpy)2(PIPNH)]2+, have been synthesized and characterized. Gel retardation assay, atomic force microscopy, and dynamic light scattering studies show that both complexes can induce the condensation of originally circular plasmid pBR322 DNA to particulate structures under neutral conditions.
Co-reporter:Hui-Juan Yu, Shu-Mei Huang, Lv-Ying Li, Hai-Na Jia, Hui Chao, Zong-Wan Mao, Jian-Zhong Liu, Liang-Nian Ji
Journal of Inorganic Biochemistry 2009 Volume 103(Issue 6) pp:881-890
Publication Date(Web):June 2009
DOI:10.1016/j.jinorgbio.2009.03.005
Two new ruthenium complexes [Ru(bpy)2(mitatp)](ClO4)21 and [Ru(bpy)2(nitatp)](ClO4)22 (bpy = 2,2′-bipyridine, mitatp = 5-methoxy-isatino[1,2-b]-1,4,8,9-tetraazatriphenylene, nitatp = 5-nitro-isatino[1,2-b]-1,4,8,9-tetraazatriphenylene) have been synthesized and characterized by elemental analysis, 1H NMR, mass spectrometry and cyclic voltammetry. Spectroscopic and viscosity measurements proved that the two Ru(II) complexes intercalate DNA with larger binding constants than that of [Ru(bpy)2(dppz)]2+ (dppz = dipyrido[3,2-a:2′,3′-c]phenazine) and possess the excited lifetime of microsecond scale upon binding to DNA. Both complexes can efficiently photocleave pBR322 DNA in vitro under irradiation. Singlet oxygen (1O2) was proved to contribute to the DNA photocleavage process, the 1O2 quantum yields was determined to be 0.43 and 0.36 for 1 and 2, respectively. Moreover, a photoinduced electron transfer mechanism was also found to be involved in the DNA cleavage process.
Co-reporter:Feng Gao, Xing Chen, Feng Zhou, Li-Ping Weng, Li-Tian Guo, Meng Chen, Hui Chao, Liang-Nian Ji
Inorganica Chimica Acta 2009 Volume 362(Issue 14) pp:4960-4966
Publication Date(Web):10 November 2009
DOI:10.1016/j.ica.2009.07.034
A series of new mononuclear polypyridyl ruthenium(II) complexes containing two imidazole groups have been synthesized and characterized by 1H NMR, ES-MS, FAB-MS, IR spectra and elemental analysis. Their electrochemistry and spectroscopic properties including UV–Vis absorption, steady-state and time-resolved emission have been studied using both experimental methods and theoretical calculations. These complexes have been found to be sensitive luminescent pH switches, as their absorption and emission spectra respond to the solutions pH disturbances with extraordinary sensitivity, through the protonation and deprotonation of the imidazole groups. DFT calculations have been carried out to reveal the exact protonation and deprotonation route. The ground-state and excited-state ionization constants (pKa and pKa∗) of each acid–base equilibrium have also been calculated according to the absorbance and emission data under different pH.A series of new ruthenium(II) complexes of ligands containing two imidazole groups have been synthesized and characterized. The complexes have been found to sensitively respond to the solutions pH changes though protonation and deprotonation of the imidazole-containing ligands, serving as luminescent pH switches with extraordinary sensitivity in aqueous solution.
Co-reporter:Bin Peng, Xiang Chen, Ke-Jie Du, Bo-Le Yu, Hui Chao, Liang-Nian Ji
Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy 2009 Volume 74(Issue 4) pp:896-901
Publication Date(Web):November 2009
DOI:10.1016/j.saa.2009.08.031
A novel ligand dipyrido[1,2,5]oxadiazolo[3,4-b]quinoxaline (dpoq) and its complexes [Ru(bpy)2(dpoq)]2+ and [Ru(phen)2(dpoq)]2+ (bpy = 2,2′-bipyridine; phen = 1,10-phenanthroline) have been synthesized and characterized by elemental analysis, electrospray mass spectra and 1H NMR. The interaction of Ru(II) complexes with calf thymus DNA (CT-DNA) was investigated by absorption spectroscopy, fluorescence spectroscopy, thermal denaturation and viscosity measurements. Results suggest that two Ru(II) complexes bind to DNA via an intercalative mode.
Co-reporter:Hui-Juan Yu, Hui Chao, Long Jiang, Lv-Ying Li, Shu-Mei Huang, Liang-Nian Ji
Inorganic Chemistry Communications 2008 Volume 11(Issue 5) pp:553-556
Publication Date(Web):May 2008
DOI:10.1016/j.inoche.2008.02.008
A novel di-bithiazolyl ruthenium(II) complex [Ru(btz)2(dppz)]2+ (btz = 4,4′-bithiazole; dppz = dipyrido[3,2-a:2′,3′-c]phenazine) has been synthesized and characterized. The DNA binding behaviors of this complex have been studied by spectroscopic and viscosity measurements. Results suggest that the complex binds to CT DNA via an intercalative mode. Upon irradiation at 365 nm, this complex was found to promote the cleavage of plasmid pBR322 DNA from supercoiled form I to nicked form II. The mechanism studies reveal that singlet oxygen 1O2 plays a significant role in the photocleavage.A novel di-bithiazolyl Ru(II) complex [Ru(btz)2(dppz)]2+ has been synthesized and characterized, the complex exhibits a high DNA-binding affinity and cleaves DNA under irradiation at 365 nm.
Co-reporter:Yi-Xian Yuan, Yu Chen, Yi-Can Wang, Cheng-Yong Su, Si-Min Liang, Hui Chao, Liang-Nian Ji
Inorganic Chemistry Communications 2008 Volume 11(Issue 9) pp:1048-1050
Publication Date(Web):September 2008
DOI:10.1016/j.inoche.2008.05.026
A new ruthenium(II) polypyridine complex, [Ru(bpy)2(PAIDH)]2+ (1) (bpy = 2,2′-bipyridine; PAIDH = 2-pyridyl-1H-anthra[1,2-d]imidazole-6,11-dione) has been found to display electrochemically “off–on” luminescent switch through exchanging two protons and two electrons between the quinone/hydroquinone redox couple at room temperature.A new ruthenium(II) polypyridine complex [Ru(bpy)2(PAIDH)]2+ exhibits electrochemically “off–on” luminescent switch through exchanging two protons and two electrons between the quinone/hydroquinone redox couple at room temperature.
Co-reporter:Feng Gao, ;Liang-Nian Ji
Chemistry & Biodiversity 2008 Volume 5( Issue 10) pp:1962-1979
Publication Date(Web):
DOI:10.1002/cbdv.200890181
Abstract
The interaction of ruthenium(II)-polypyridyl complexes with DNA has attracted considerable interests during the past two decades. This paper presents some recent progresses in our laboratory on the interaction of RuII-polypyridyl complexes with DNA. The first part describes the effect of modulating the intercalative ligand on the DNA-binding behaviors of the complexes, such as DNA-binding affinity, DNA-binding enantioselectivity, DNA molecular ‘light switch’ effect, and DNA sequence selectivity. The second part focuses on the DNA photocleavage by the complexes and its mechanism. In the final part, we discuss the topoisomerase inhibition and its mechanism, as well as the antitumor activity of the RuII-polypyridyl complexes.
Co-reporter:Feng Gao;Yuan-Fang Wei;Yi-Xian Yuan;Bin Peng;Xin Chen;Kang-Cheng Zheng;Liang-Nian Ji
Helvetica Chimica Acta 2008 Volume 91( Issue 3) pp:395-410
Publication Date(Web):
DOI:10.1002/hlca.200890045
Abstract
Two new complexes, [Ru(phen)2(ppd)]2+ (1) and [Ru(phen)(ppd)2]2+ (2) (ppd=pteridino[6,7-f] [1,10]phenanthroline-11,13(10H,12H)-dione, phen=1,10-phenanthroline) were synthesized and characterized by ES-MS, 1H-NMR spectroscopy, and elemental analysis. The intercalative DNA-binding properties of 1 and 2 were investigated by absorption-spectroscopy titration, luminescence-spectroscopy studies, thermal denaturation, and viscosity measurements. The theoretical aspects were further discussed by comparative studies of 1 and 2 by means of DFT calculations and molecular-orbital theory. Photoactivated cleavage of pBR322 DNA by the two complexes were also studied, and 2 was found to be a much better photocleavage reagent than 1. The mechanism studies revealed that singlet oxygen and the excited-states redox potentials of the complex may play an important role in the DNA photocleavage.
Co-reporter:Bin Peng, Hui Chao, Bin Sun, Hong Li, Feng Gao, Liang-Nian Ji
Journal of Inorganic Biochemistry 2007 Volume 101(Issue 3) pp:404-411
Publication Date(Web):March 2007
DOI:10.1016/j.jinorgbio.2006.11.008
Two novel cobalt(III) mixed-polypyridyl complexes [Co(phen)2(dpta)]3+ and [Co(phen)2(amtp)]3+ (phen = 1,10-phenanthroline, dpta = dipyrido-[3,2-a;2′,3′-c]- thien-[3,4-c]azine, amtp = 3-amino-1,2,4-triazino[5,6-f]1,10-phenanthroline) have been synthesized and characterized. The interaction of these complexes with calf thymus DNA was investigated by spectroscopic, cyclic voltammetry, and viscosity measurements. Results suggest that the two complexes bind to DNA via an intercalative mode. Moreover, these Co(III) complexes have been found to promote the photocleavage of plasmid DNA pBR322 under irradiation at 365 nm. The mechanism studies reveal that hydroxyl radical (OH) is likely to be the reactive species responsible for the cleavage of plasmid DNA by [Co(phen)2(dpta)]3+ and superoxide anion radical (O2-) acts as the key role in the cleavage reaction of plasmid DNA by [Co(phen)2(amtp)]3+.
Co-reporter:Feng Gao;Feng Zhou;Lian-Cai Xu;Kang-Cheng Zheng;Liang-Nian Ji
Helvetica Chimica Acta 2007 Volume 90(Issue 1) pp:36-51
Publication Date(Web):26 JAN 2007
DOI:10.1002/hlca.200790019
Two novel chiral ruthenium(II) complexes, Δ-[Ru(bpy)2(dmppd)]2+ and Λ-[Ru(bpy)2(dmppd)]2+ (dmppd = 10,12-dimethylpteridino[6,7-f] [1,10]phenanthroline-11,13(10H,12H)-dione, bpy = 2,2′-bipyridine), were synthesized and characterized by elemental analysis, 1H-NMR and ES-MS. The DNA-binding behaviors of both complexes were studied by UV/VIS absorption titration, competitive binding experiments, viscosity measurements, thermal DNA denaturation, and circular-dichroism spectra. The results indicate that both chiral complexes bind to calf-thymus DNA in an intercalative mode, and the Δ enantiomer shows larger DNA affinity than the Λ enantiomer does. Theoretical-calculation studies for the DNA-binding behaviors of these complexes were carried out by the density-functional-theory method. The mechanism involved in the regulating and controlling of the DNA-binding abilities of the complexes was further explored by the comparative studies of [Ru(bpy)2(dmppd)]2+ and of its parent complex [Ru(bpy)2(ppd)]2+ (ppd = pteridino[6,7-f] [1,10]phenanthroline-11,13 (10H,12H)-dione).
Co-reporter:Feng Gao;Jin-Quan Wang
JBIC Journal of Biological Inorganic Chemistry 2007 Volume 12( Issue 7) pp:1015-1027
Publication Date(Web):2007 September
DOI:10.1007/s00775-007-0272-4
Many antitumor drugs act as topoisomerase inhibitors, and the inhibitions are usually related to DNA binding. Here we designed and synthesized DNA-intercalating Ru(II) polypyridyl complexes Δ--[Ru(bpy)2(uip)]2+ and Λ-[Ru(bpy)2(uip)]2+ (bpy is 2,2′-bipyridyl, uip is 2-(5-uracil)-1H-imidazo[4,5-f][1,10]phenanthroline). The DNA binding, photocleavage, topoisomerase inhibition, and cytotoxicity of the complexes were studied. As we expected, the synthesized Ru(II) complexes can intercalate into DNA base pairs and cleave the pBR322 DNA with high activity upon irradiation. The mechanism studies reveal that singlet oxygen (1O2) and superoxide anion radical (O2•−) may play an important role in the photocleavage. The inhibition of topoisomerases I and II by the Ru(II) complexes has been studied. The results suggest that both complexes are efficient inhibitors towards topoisomerase II by interference with the DNA religation and direct topoisomerase II binding. Both complexes show antitumor activity towards HELA, hepG2, BEL-7402, and CNE-1 tumor cells.
Co-reporter:Feng Gao, Hui Chao, Feng Zhou, Yi-Xian Yuan, Bin Peng, Liang-Nian Ji
Journal of Inorganic Biochemistry 2006 Volume 100(Issue 9) pp:1487-1494
Publication Date(Web):September 2006
DOI:10.1016/j.jinorgbio.2006.04.008
A novel polypyridyl ligand pteridino[7,6-f][1,10]phenanthroline-1,13(10H,12H)-dione (ppd) and its ruthenium(II) complex [Ru(bpy)2ppd]2+ have been synthesized and characterized by elemental analysis, electrospray mass spectra and 1H NMR. The interaction of the complex with calf thymus DNA was investigated by spectroscopic and viscosity measurements. The results suggest that the complex binds to DNA via an intercalative mode and serves as a molecular “light switch” for DNA. Moreover, the complex has been found to promote the photocleavage of plasmid DNA pBR322 under irradiation at 365 nm. The mechanism studies reveal that singlet oxygen (1O2) and superoxide anion radical (O2-) play a significant role in the photocleavage.
Co-reporter:Yun-Jun Liu, Hui Chao, Yi-Xian Yuan, Hui-Juan Yu, Liang-Nian Ji
Inorganica Chimica Acta 2006 Volume 359(Issue 12) pp:3807-3814
Publication Date(Web):1 September 2006
DOI:10.1016/j.ica.2006.05.015
Two novel Ru(II) complexes [Ru(bpy)2(MCMIP)]2+ (1) and [Ru(phen)2(MCMIP)]2+ (2) (bpy = 2,2′-bipyridine; phen = 1,10-phenanthroline; MCMIP = 2-(6-methyl-3-chromonyl)imidazo[4,5-f][1,10]-phenanthroline) have been synthesized and characterized by elemental analysis, mass spectra and 1H NMR. The DNA-binding properties of the complexes were investigated by absorption, emission, melting temperature and viscosity measurements. Experimental results indicate that the two complexes can intercalate into DNA base pairs. Upon irradiation at 365 nm, two Ru(II) complexes were found to promote the cleavage of plasmid pBR 322 DNA from supercoiled form I to nicked form II, and the mechanisms for DNA cleavage by the complexes were also investigated.Two novel ruthenium(II) complexes [Ru(bpy)2(MCMIP)]2+ (1) and [Ru(phen)2(MCMIP)]2+ (2) have been synthesized and characterized. Their DNA-binding and photocleavage properties have also been investigated by spectroscopic methods, viscosity and electrophoresis measurements.
Co-reporter:Huaiyi Huang, Liang Yang, Pingyu Zhang, Kangqiang Qiu, Juanjuan Huang, Yu Chen, JiaJie Diao, Jiankang Liu, Liangnian Ji, Jiangang Long, Hui Chao
Biomaterials (March 2016) Volume 83() pp:321-331
Publication Date(Web):March 2016
DOI:10.1016/j.biomaterials.2016.01.014
Mitochondrial fission and fusion control the shape, size, number, and function of mitochondria in the cells of organisms from yeast to mammals. The disruption of mitochondrial fission and fusion is involved in severe human diseases such as Parkinson's disease, Alzheimer's disease, metabolic diseases, and cancers. Agents that can real-time track the mitochondrial dynamics are of great importance. However, the short excitation wavelengths and rapidly photo-bleaching properties of commercial mitochondrial dyes render them unsuitable for tracking mitochondrial dynamics. Thus, mitochondrial targeting agents that exhibit superior photo-stability under continual light irradiation, deep tissue penetration and at intrinsically high three-dimensional resolutions are urgently needed. Two-photon-excited compounds employ low-energy near-infrared light and have emerged as a non-invasive tool for real-time cell imaging. Here, cyclometalated Ir(III) complexes (Ir1–Ir5) are demonstrated as one- and two-photon phosphorescent probes for the real-time imaging and tracking of mitochondrial fission and fusion. The results indicate that Ir2 is well suited for two-photon phosphorescent tracking of mitochondrial fission and fusion in living cells and in Caenorhabditis elegans (C. elegans). This study provides a practical use for mitochondrial targeting two-photon phosphorescent Ir(III) complexes.
Co-reporter:Huaiyi Huang, Liang Yang, Pingyu Zhang, Kangqiang Qiu, Juanjuan Huang, Yu Chen, JiaJie Diao, Jiankang Liu, Liangnian Ji, Jiangang Long, Hui Chao
Biomaterials (March 2016) Volume 83() pp:321-331
Publication Date(Web):March 2016
DOI:10.1016/j.biomaterials.2016.01.014
Co-reporter:Kai Xiong, Yu Chen, Cheng Ouyang, Rui-Lin Guan, Liang-Nian Ji, Hui Chao
Biochimie (June 2016) Volume 125() pp:
Publication Date(Web):June 2016
DOI:10.1016/j.biochi.2016.03.013
•Ir1–Ir4 possess selectivity between human cancer and normal cells.•Ir1–Ir4 enter cancer cells and normal cells via different mechanisms.•Ir4 shown much more potential than Ir2 and Ir3, even though they are isomers.•Treatment of Ir1–Ir4 results in generation of ROS and prominent depletion of Δψm.Four cyclometalated iridium(III) complexes [Ir(dfppy)2(L)]+ (dfppy = 2-(2,4-difluorophenyl)pyridine, L = 6-(pyridin-2-yl)-1,3,5-triazine-2,4-diamine, Ir1; 6-(isoquinolin-1-yl)-1,3,5-triazine-2,4-diamine, Ir2; 6-(quinolin-2-yl)-1,3,5-triazine-2,4-diamine, Ir3; 6-(isoquinolin-3-yl)-1,3,5-triazine-2,4-diamine, Ir4) have been synthesized and characterized. Distinct from cisplatin, Ir1–Ir4 could specifically target mitochondria and induced apoptosis against various cancer cell lines, especially for cisplatin resistant cells. ICP-MS results indicated that Ir1–Ir4 were taken up via different mechanism for cancer cells and normal cells, which resulted in their high selectivity. The structure-activity relationship and signaling pathways were also discussed.
Co-reporter:Bin Sun, Jun Chu, Yu Chen, Feng Gao, Liang-Nian Ji, Hui Chao
Journal of Molecular Structure (12 November 2008) Volume 890(Issues 1–3) pp:203-208
Publication Date(Web):12 November 2008
DOI:10.1016/j.molstruc.2008.05.035
Co-reporter:Pingyu Zhang, Jinquan Wang, Huaiyi Huang, Kangqiang Qiu, Juanjuan Huang, Liangnian Ji and Hui Chao
Journal of Materials Chemistry A 2017 - vol. 5(Issue 4) pp:NaN678-678
Publication Date(Web):2016/11/29
DOI:10.1039/C6TB01991A
Gold nanorods (AuNRs) and nanostars (AuNTs) were widely applied in photothermal cancer therapy recently. However, due to the photothermal effect, naked AuNRs and AuNTs easily melt into gold spheres. This drawback results in loss of the characteristic near-infrared (NIR) surface plasmon resonance (SPR) and limits their therapeutic applications. In this paper, we reported that ruthenium(II) complex-functionalized AuNRs (AuNRs@Ru) and AuNTs (AuNTs@Ru) exhibit higher photothermal stability and photothermal efficiency than naked AuNRs and AuNTs. AuNRs@Ru and AuNTs@Ru maintain the morphology and NIR SPR absorption of gold nanoparticles upon 0.25 W cm−2 laser irradiation, which is lower than the maximal permissible exposure of skin as per ANSI regulation (0.33 W cm−2 at 808 nm). Further photothermal therapy studies on three-dimensional (3D) HeLa spheroids and an in vivo tumor model show that AuNRs@Ru and AuNTs@Ru are more effective for the photothermal destruction of tumors than AuNRs and AuNTs.
Co-reporter:Kangqiang Qiu, Yukang Liu, Huaiyi Huang, Chaofeng Liu, Hongyi Zhu, Yu Chen, Liangnian Ji and Hui Chao
Dalton Transactions 2016 - vol. 45(Issue 41) pp:NaN16147-16147
Publication Date(Web):2016/09/21
DOI:10.1039/C6DT03328H
An efficient method was developed that controls biscylometalated iridium(III) complexes to target mitochondria or lysosomes by regulating the lipophilicity of the main ligands.
Co-reporter:Kejie Du, Jiewen Liang, Yi Wang, Junfeng Kou, Chen Qian, Liangnian Ji and Hui Chao
Dalton Transactions 2014 - vol. 43(Issue 46) pp:NaN17316-17316
Publication Date(Web):2014/09/24
DOI:10.1039/C4DT02142H
Five novel ruthenium(II) complexes, [Ru(dtzp)(dppt)]2+ (1), [Ru(dtzp)(pti)]2+ (2), [Ru(dtzp)(ptn)]2+ (3), [Ru(dtzp)(pta)]2+ (4) and [Ru(dtzp)(ptp)]2+ (5) (where dtzp = 2,6-di(thiazol-2-yl)pyridine, dppt = 3-(1,10-phenanthroline-2-yl)-5,6-diphenyl-as-triazine), pti = 3-(1,10-phenanthroline-2-yl)-as-triazino-[5,6-f]isatin, ptn = 3-(1,10-phenanthroline-2-yl)-as-triazino[5,6-f]naphthalene, pta = 3-(1,10-phenanthroline-2-yl)-as-triazino[5,6-f]acenaphthylene, and ptp = 3-(1,10-phenanthroline-2-yl)-as-triazino[5,6-f]-phenanthrene), were synthesised and characterised. The structures of complexes 3–5 were determined by X-ray diffraction. The DNA binding behaviours of the complexes were studied by spectroscopic and viscosity measurements. The results suggested that the Ru(II) complexes, except for complex 1, bind to DNA in an intercalative mode. Topoisomerase inhibition and DNA strand passage assay confirmed that Ru(II) complexes 3, 4, and 5 acted as efficient dual inhibitors of topoisomerases I and IIα. In vitro cytotoxicity assays indicated that these complexes exhibited anticancer activity against various cancer cell lines. Ruthenium(II) complexes were confirmed to preferentially accumulate in the nucleus of cancer cells and induced DNA damage. Flow cytometric analysis and AO/EB staining assays indicated that these complexes induced cell apoptosis. With the loss of the mitochondrial membrane potential, the Ru(II) complexes induce apoptosis via the mitochondrial pathway.
Co-reporter:Miao Ouyang, Leli Zeng, Huaiyi Huang, Chengzhi Jin, Jiangping Liu, Yu Chen, Liangnian Ji and Hui Chao
Dalton Transactions 2017 - vol. 46(Issue 20) pp:NaN6744-6744
Publication Date(Web):2017/05/01
DOI:10.1039/C7DT01043E
Six cyclometalated iridium(III) complexes bearing different numbers of fluorine atoms were synthesized. These complexes demonstrated much better anti-proliferation activities towards five tumour cell lines than the widely used clinical chemotherapeutic agent cisplatin. Moreover, the anti-proliferation activities were correlated to the number of substituted fluorine atoms. Colocalization and inductively coupled plasma-mass spectrometry (ICP-MS) indicated that this series of complexes could penetrate cell membranes rapidly and preferentially target mitochondria. Manifesting high selectivity between tumour cells and normal cells and remarkable sensitivity to a cisplatin-resistant cell line (A549R), complex Ir6 was successfully developed as a novel anticancer agent (with IC50 values of 0.5 ± 0.1 μM for HeLa, 1.1 ± 0.2 μM for HepG2, 1.5 ± 0.3 μM for BEL-7402, 0.8 ± 0.1 μM for A549, and 0.7 ± 0.2 μM for A549R cell lines). Further mechanism studies including mitochondrial membrane potential depolarization and caspase 3/7 activation revealed that Ir6 induced apoptosis via mitochondrial pathways. These results demonstrated that complex Ir6 might be a promising candidate as a mitochondria-targeted theranostic anticancer agent.
Co-reporter:Bole Yu, Ying Chen, Mei Hong, Pingping Duan, Shifeng Gan, Hui Chao, Zujin Zhao and Jing Zhao
Chemical Communications 2015 - vol. 51(Issue 76) pp:NaN14368-14368
Publication Date(Web):2015/08/03
DOI:10.1039/C5CC05239D
Using an internally oxidizing directing group (DG) strategy, we report a RhIII-catalyzed synthesis of isoquinolones via C–H activation/annulation of benzoylhydrazines and alkynes. Tunable double cascade cyclization of benzoylhydrazines with two equivalents of alkynes led to tetracyclic amides. These N-heterocycles demonstrated adjustable AIE properties.
Co-reporter:Kangqiang Qiu, Miao Ouyang, Yukang Liu, Huaiyi Huang, Chaofeng Liu, Yu Chen, Liangnian Ji and Hui Chao
Journal of Materials Chemistry A 2017 - vol. 5(Issue 27) pp:NaN5498-5498
Publication Date(Web):2017/06/15
DOI:10.1039/C7TB00731K
By integrating targeting, imaging and treatment, organelle-targeted photodynamic therapy (PDT) has been reported to be an effective strategy for cancer therapy. However, targeting leads to the accumulation of photosensitizers (PSs) in the targeted organelles, which leads to a reduction in 1O2 generation and fluorescence quenching, especially for the lipophilic mitochondria-targeted PSs. Moreover, because PSs always need exposure to light for a specific period, photobleaching is difficult to avoid. To address these issues, two iridium(III) complexes with aggregation-induced two-photon emission (AITPE) characteristics were developed. With lipophilicity, the complexes aggregated in water and targeted mitochondria. Owing to their impressive 1O2 production quantum yields and excellent two-photon properties in the aggregate states, the complexes were successfully used for mitochondria-targeted two-photon PDT in monolayer cells and multicellular spheroids. Our results highlighted that the use of a PS with aggregation enhanced 1O2 generation and fluorescence is an effective solution for aggregation in organelle-targeted PDT.
Co-reporter:Pingyu Zhang, Jinquan Wang, Huaiyi Huang, Liping Qiao, Liangnian Ji and Hui Chao
Dalton Transactions 2013 - vol. 42(Issue 24) pp:NaN8917-8917
Publication Date(Web):2013/04/18
DOI:10.1039/C3DT50472G
A series of novel chiral ruthenium(II) complexes with phenolic hydroxyl groups were synthesized and characterized. These ruthenium(II) complexes exhibited strong dual inhibition of topoisomerases I and IIα, with approximate IC50 values of 3–15 mM, which were more efficient than the widely clinically used single TopoI poison camptothecin (CPT) or TopoIIα poison etoposide (VP-16). Δ-1 and Λ-1 with more hydroxyls were observed to be more potent inhibitors. To further evaluate the mechanism of the complexes at a cellular level, these complexes were investigated for their effect on cell proliferation, cell cycle progression and induction of apoptosis. The results indicated that ruthenium(II) complexes permeated the nuclei in cancer cells and inhibited the activities of nuclear enzymes topoisomerases I and IIα, then triggered DNA damage and induced apoptosis in the cancer cells. The simultaneous inhibition of TopoI and TopoIIα induced the death of cancer cells, which may be a promising and effective strategy for cancer therapy.
Co-reporter:Guoliang Liao, Xiang Chen, Jingheng Wu, Chen Qian, Yi Wang, Liangnian Ji and Hui Chao
Dalton Transactions 2015 - vol. 44(Issue 34) pp:NaN15156-15156
Publication Date(Web):2015/01/07
DOI:10.1039/C4DT03585B
One novel ruthenium polypyridyl complex, [Ru(bpy)2(icip)]2+ (1), and two previously reported ruthenium polypyridyl complexes, [Ru(bpy)2(pdppz)]2+ (2) and [Ru(bpy)2(tactp)]2+ (3) (bpy = 2,2′-bipyridine, icip = 2-(indeno[2,1-b]chromen-6-yl)-1H-imidazo[4,5-f][1,10]phenanthroline, pdppz = phenanthro[4,5-abc]dipyrido[3,2-h:2′,3′-j]phenazine, tactp = 4,5,9,18-tetraazachryseno[9,10-b]-triphenylene), have been synthesised. As expected, these complexes show inhibition towards telomerase by inducing and stabilising the G-quadruplex structure, and behave as topoisomerase I/II poisons at the same time. Additionally, the acute and chronic cytotoxicities of the complexes are considered. Furthermore, cell apoptosis experiments are used to briefly study the mechanism. Because studies involving multi-target inhibition towards topoisomerase and telomerase of Ru(II) complexes have not been reported previously, the present research may help to develop innovative chemical strategies and therapies.
Co-reporter:Yu Chen, Liping Qiao, Bole Yu, Guanying Li, Chunyuan Liu, Liangnian Ji and Hui Chao
Chemical Communications 2013 - vol. 49(Issue 94) pp:NaN11097-11097
Publication Date(Web):2013/10/08
DOI:10.1039/C3CC46957C
An AIPE-active iridium(III) complex was found to possess high specificity for mitochondria, superior photostability, low cytotoxicity, and high resistance to the loss of mitochondrial membrane potential. Thus, this complex can be used for mitochondrial imaging and tracking in living cells.
Co-reporter:Xiang Chen, Jing-Heng Wu, Ying-Wei Lai, Rong Zhao, Hui Chao and Liang-Nian Ji
Dalton Transactions 2013 - vol. 42(Issue 13) pp:NaN4397-4397
Publication Date(Web):2013/01/10
DOI:10.1039/C3DT32921F
Two ruthenium(II) polypyridyl complexes, [Ru(bpy)2(ptpn)]2+ (1) (bpy = 2,2′-bipyridine, ptpn = 3-(1,10-phenanthroline-2-yl)-as-triazino[5,6-f]1,10-phenanthroline) and [Ru(phen)2(ptpn)]2+ (2) (phen = 1,10-phenanthroline), were synthesized and characterized. Crystal structure analysis shows that complex 1 has a large planar aromatic area and possesses the potential to fit the geometric structure of G-quadruplex. The interaction of the G-quadruplex DNA with Ru(II) complexes was explored by means of circular dichroism (CD), fluorescence resonance energy transfer (FRET) melting assay, competitive FRET assay and polymerase chain reaction (PCR) stop assay. The results indicated that complexes 1 and 2 both have the ability to promote the formation and stabilization of the human telomeric d[(TTAGGG)n] (HTG22) quadruplex and exhibit high G-quadruplex DNA selectivity over duplex DNA. The telomere repeat amplification protocol (TRAP) assay and long-term proliferation experiments further demonstrate that the Ru(II) complexes are potent telomerase inhibitors and HeLa cell proliferation inhibitors.
Co-reporter:Guanying Li, Yu Chen, Jinquan Wang, Qian Lin, Jing Zhao, Liangnian Ji and Hui Chao
Chemical Science (2010-Present) 2013 - vol. 4(Issue 12) pp:NaN4433-4433
Publication Date(Web):2013/09/02
DOI:10.1039/C3SC52301B
A non-emissive dinuclear iridium(III) complex (SP-2) linked via an azo group has been adopted as a phosphorescent probe for sulphite and bisulphite. SP-2 exhibits a turn-on phosphorescent response toward sulphite and bisulphite with high selectivity and sensitivity. Furthermore, this probe can be used for living cell imaging to identify the presence of both external sulphite/bisulphite supplemented into cell cultures and endogenous sulphite/bisulphite biosynthesised in living cells.
Co-reporter:Bole Yu, Yu Chen, Cheng Ouyang, Huaiyi Huang, Liangnian Ji and Hui Chao
Chemical Communications 2013 - vol. 49(Issue 8) pp:NaN812-812
Publication Date(Web):2012/12/06
DOI:10.1039/C2CC37896E
A luminescent tetranuclear ruthenium(II) complex was developed to act as a DNA carrier and at the same time offer luminescent imaging to follow the DNA intracellular trafficking with time.
Co-reporter:Kangqiang Qiu, Huaiyi Huang, Bingyang Liu, Yukang Liu, Pingyu Zhang, Yu Chen, Liangnian Ji and Hui Chao
Journal of Materials Chemistry A 2015 - vol. 3(Issue 32) pp:NaN6697-6697
Publication Date(Web):2015/07/20
DOI:10.1039/C5TB01091H
Two-photon phosphorescent probes have emerged as promising molecular tools for imaging subcellular organelles. Here, the facile synthesis of four new iridium(III)-based mitochondrial probes with two-photon phosphorescence, Ir1–Ir4, is presented. Ir1–Ir4 possess high specificity for mitochondrial localization, which is advantageous in comparison with commercially available mitochondrial trackers of changes in the mitochondrial membrane potential in live cells. In addition to low cytotoxicity and high resistance to photobleaching, Ir1–Ir4 are applicable for imaging and tracking of mitochondrial morphological changes during the early stages of apoptosis. While naturally possessing intensive two-photon properties, Ir1–Ir4 were further developed for imaging of the mitochondria in 3D multicellular spheroids.
Co-reporter:Guoliang Liao, Xiang Chen, Jingheng Wu, Chen Qian, Hanqiang Wang, Liangnian Ji and Hui Chao
Dalton Transactions 2014 - vol. 43(Issue 21) pp:NaN7819-7819
Publication Date(Web):2014/03/10
DOI:10.1039/C3DT53547A
Two novel ruthenium polypyridyl complexes, Ru[(bpy)2(pedppz)]2+ (1) and Ru[(bpy)2(pemitatp)]2+ (2) (bpy = 2′2-bipyridine, pdeppz = 10-(2-(piperidin-1-yl)ethoxy)dipyrido[3,2-a:2′,3′-c]phenazine, pemitatp = 5-methoxy-1-(2-(piperidin-1-yl)ethyl)-isatino[1,2-b]-1,4,8,9-tetraazatriphenylene), bearing large planar π-delocalized aromatic systems with flexible chains have been synthesised and characterised. As expected, these complexes show inhibition towards telomerase by inducing and stabilising the G-quadruplex structure.
Co-reporter:Guanying Li, Qian Lin, Liangnian Ji and Hui Chao
Journal of Materials Chemistry A 2014 - vol. 2(Issue 45) pp:NaN7926-7926
Publication Date(Web):2014/09/18
DOI:10.1039/C4TB01251H
Mitochondria are one of the major sources of cellular reactive oxygen species (ROS) and the hypochlorite ion (ClO−) is one kind of highly ROS involved in many important biological events. Herein, we present a novel class of mitochondrial ClO− probes. A series of iridium(III) complexes, Ir1–Ir4, were synthesized which exhibited a highly specific response of their phosphorescence signal toward ClO− over other ROS. By changing the cyclometalated ligands, the turn-on phosphorescent emission colours can be tuned from green to red. ICP-MS results and Mito-tracker co-staining experiments revealed that Ir1–Ir4 localized specifically in mitochondria. Finally, complexes Ir1–Ir4 were successfully applied in subcellular imaging for detection of endogenous ClO− in mitochondria.
Co-reporter:Chen Qian, Jingheng Wu, Liangnian Ji and Hui Chao
Dalton Transactions 2016 - vol. 45(Issue 26) pp:NaN10555-10555
Publication Date(Web):2016/05/13
DOI:10.1039/C6DT01422D
Four chiral Ru(II) complexes bearing furan ligands, Δ/Λ-[Ru(bpy)2(pocl)]2+ (Δ/Λ-1) and Δ/Λ-[Ru(bpy)2(poi)]2+ (Δ/Λ-2) (bpy = 2,2′-bipyridine, pocl = 2-(5-chlorofuran-2-yl)imidazo[4,5-f][1,10]phenanthroline, poi = 2-(5-5-iodofuran-2-yl)imidazo[4,5-f][1,10]phenanthroline), were synthesized and characterized. These Ru(II) complexes showed antitumor activities against HeLa, A549, HepG2, HL-60 and K562 tumor cell lines, especially the HL-60 tumor cell line. Moreover, Δ-2 was more active than other complexes accounting for the different cellular uptakes. In addition, Δ-2 could accumulate in the nucleus of HL-60 cells, suggesting that nucleic acids were the cellular target of Δ-2. Topoisomerase inhibition tests in vitro and in living cells confirmed that the four complexes acted as efficient topoisomerase IIα poisons, DNA double-strand breaks had also been observed from neutral single cell gel electrophoresis (comet assay). Δ-2 inhibited the growth of HL-60 cells through the induction of apoptotic cell death, as evidenced by the Alexa Fluor® 488 annexin V staining assays. The results demonstrated that Δ-2 acted as a topoisomerase IIα poison and caused DNA double-strand damage that could lead to apoptosis.
Co-reporter:Huaiyi Huang, Pingyu Zhang, Bole Yu, Chengzhi Jin, Liangnian Ji and Hui Chao
Dalton Transactions 2015 - vol. 44(Issue 39) pp:NaN17345-17345
Publication Date(Web):2015/09/01
DOI:10.1039/C5DT02081F
This study investigated the photodynamic therapy (PDT) and anticancer activity of mixed ligand Ru(II) terpyridyl complexes (Ru1–Ru3). The photophysical and photochemical properties, hydrophobic properties, DNA binding and DNA transcription inhibition abilities, cell uptake efficiency, cellular localization and photo-cytotoxicity were investigated. Ru1–Ru3 exhibited red luminescence between 670–710 nm and functioned as photo-sensitizers (PSs) by generating both singlet oxygen and radical ions. Without light activation, Ru1–Ru3 were located at the cytoplasm and were nontoxic to cells. However, upon light activation, Ru1–Ru3 exhibited significant photocytotoxicity. After PDT treatment, mitochondria alteration and nuclear membrane disruption occurred, which resulted in relocalization of the complexes from the cytoplasm to the nucleus. Moreover, high cellular oxidative stress caused cell necrocytosis after PDT treatment.
Co-reporter:Guanying Li, Lingli Sun, Liangnian Ji and Hui Chao
Dalton Transactions 2016 - vol. 45(Issue 34) pp:NaN13276-13276
Publication Date(Web):2016/07/11
DOI:10.1039/C6DT01624C
The DNA photoswitch [Ru(bpy)2dppz]2+ (bpy = 2,2′-bipyridine, dppz = dipyrido[3,2-a:2′,3′-c]phenazine) has attracted much attention and become a powerful tool for studying the interaction of metal polypyridyl complexes with DNA. A large number of Ru-dppz complexes have been designed for a wide range of uses in many fields. In this perspective, we first introduce the latest results of Ru-dppz complexes that bind with DNA. The mechanisms of the light-switch effect and the structural modifications of Ru-dppz systems are also briefly introduced. We also review the recent advances in biological applications of the Ru-dppz system in DNA binders, cellular imaging, anticancer drugs, protein aggregation detection and chemosensors.
Co-reporter:Huaiyi Huang, Pingyu Zhang, Yu Chen, Liangnian Ji and Hui Chao
Dalton Transactions 2015 - vol. 44(Issue 35) pp:NaN15610-15610
Publication Date(Web):2015/07/23
DOI:10.1039/C5DT02446C
Ruthenium complexes have been considered as promising substitutes for cisplatin in cancer chemotherapy. However, novel ruthenium-based therapies are faced with some limitations, such as unimpressive cytotoxicity toward solid tumors. Herein, we designed and synthesized phenyl-substituted terpyridyl ruthenium(II) complexes ([Ru(tpy)(bpy)Cl]+ (Ru1), [Ru(phtpy)(bpy)Cl]+ (Ru2) and [Ru(biphtpy)(bpy)Cl]+ (Ru3)) which exhibited distinctly different anticancer activity. Ru1–Ru3 all underwent moderate aquation in buffer solution and this process was significantly inhibited by high chloride concentration. Cancer cells were found to readily uptake the relatively hydrophobic Ru3, as quantified using inductively coupled plasma mass spectrometry (ICP-MS). Ru1 was found to be non-cytotoxic (IC50 > 100 μM) while Ru3 exhibited very promising cytotoxicity on both two-dimensional (2D) cancer cell monolayers and 3D MCTSs. An antiproliferative assay revealed that Ru3 significantly inhibited cellular DNA replication which ultimately induced apoptosis of cancer cells.
Co-reporter:Lüying Li, Kejie Du, Yi Wang, Haina Jia, Xiaojuan Hou, Hui Chao and Liangnian Ji
Dalton Transactions 2013 - vol. 42(Issue 32) pp:NaN11588-11588
Publication Date(Web):2013/06/04
DOI:10.1039/C3DT50395J
Three mononuclear copper complexes [Cu(PDTP)Cl2] (PDTP = 4-phenyl-2,6-di(thiazole-2-yl)pyridine, CuPDTP), [Cu(ADTP)Cl2] (ADTP = 4-(anthracen-9-yl)-2,6-di(thiazole-2-yl)pyridine, CuADTP) and [Cu(BFDTP)Cl2] (BFDTP = 4-(benzofuran-2-yl)-2,6-di(thiazole-2-yl)pyridine, CuBFDTP) were synthesized and characterized. The X-ray single crystallography results indicated that the Cu(II) ions showed slightly distorted square pyramid coordination environments, and the ligands deviated from ideal planarity in all three compounds. Based on the DNA binding studies, it was demonstrated that these three complexes exhibited weak DNA binding strengths, which were most likely groove binding modes. CuPDTP, CuADTP and CuBFDTP induced efficient DNA cleavage in the dark without the addition of external catalysts (oxidant or reductant). In contrast, in the presence of reducing or oxidizing agents, the nuclease activities increased more than 10-fold. Mechanistic investigations revealed the participation of reactive oxygen species, which can be trapped by ROS radical scavengers and ROS sensors. In the same experimental conditions, the free ligands and CuCl2 did not display any DNA cleaving activity. This result indicates that the complexes, rather than their components, play a significant role in the nuclease reaction process and that DNA cleavage may be initiated in an oxidative pattern. The proposed mechanism was attributed to the in situ activation of molecular oxygen by the oxidation of the copper complexes. In the MTT cytotoxicity studies, the three Cu(II) complexes exhibited an antitumor activity against the HeLa, BEL-7402 and HepG2 tumor cell lines. The HeLa cells treated with Cu(II) complexes demonstrated marked changes in their nuclear morphology, which were detected by Hoechst 33258 nuclear staining and acridine orange/ethidium bromide (AO/EB) staining assays. Nuclear chromatin cleavage also was observed from alkaline single-cell gel electrophoresis (comet assay).
Co-reporter:Yu Chen, Wenchao Xu, Jiarui Zuo, Liangnian Ji and Hui Chao
Journal of Materials Chemistry A 2015 - vol. 3(Issue 16) pp:NaN3314-3314
Publication Date(Web):2015/03/03
DOI:10.1039/C5TB00251F
Mitochondrial dynamic processes are essential for mammalian development and are engaged in several diseases. However, commercial mitochondrial probes are currently too limited to satisfactorily track these dynamics. In this present work, seven dinuclear iridium(III) complexes [(ppy)2Ir(L1–7)Ir(ppy)2]2+ (Ir1–Ir7, ppy = 2-phenylpyridine, L = 1,3-bis(1-substituted-imidazo[4,5-f][1,10]phenanthroline-2-yl)benzene) were synthesized. Possessing a high specificity for mitochondria, low cytotoxicity and high resistance to the loss of the mitochondrial membrane potential, Ir2–Ir7 can serve as mitochondrial imaging dyes. Ir3 and Ir6 are further developed for tracking mitochondrial morphological changes during the early stages of apoptosis. In addition, the relationship of the electron donating/withdrawing groups in these systems with the photophysical properties and photostability is also discussed.
Co-reporter:Chengzhi Jin, Jiangping Liu, Yu Chen, Guanying Li, Ruilin Guan, Pingyu Zhang, Liangnian Ji and Hui Chao
Dalton Transactions 2015 - vol. 44(Issue 16) pp:NaN7547-7547
Publication Date(Web):2015/03/18
DOI:10.1039/C5DT00467E
A new series of cyclometalated iridium(III) complexes with imidazo[4,5-f][1,10]phenanthroline derivatives (i.e., MitoIr1–MitoIr7) were synthesized and developed to image mitochondria in living cells. In comparison with commercially available mitochondrial trackers, these complexes exhibit a superior capacity to selectively accumulate in mitochondria with no requirement of any membrane permeabilization or replacement of the culture medium. In addition, the excellent photostability under continuous laser irradiation as well as the stable physiological pH resistance of these complexes were confirmed by photobleaching experiments and luminescence measurements. Importantly, MitoIr7, which exhibited both excellent luminescence and high ability to locate in mitochondria, was developed to track the mitochondrial morphological changes over a long period of time.
Co-reporter:Wenchao Xu, Jiarui Zuo, Lili Wang, Liangnian Ji and Hui Chao
Chemical Communications 2014 - vol. 50(Issue 17) pp:NaN2125-2125
Publication Date(Web):2014/01/02
DOI:10.1039/C3CC48916G
A new series of dinuclear ruthenium(II) polypyridyl complexes, which possess larger π-conjugated systems, good water solubility and pH resistance, and high photostability, were developed to act as single and two-photon luminescence cellular imaging probes.
Co-reporter:Guanying Li, Yu Chen, Jingheng Wu, Liangnian Ji and Hui Chao
Chemical Communications 2013 - vol. 49(Issue 20) pp:NaN2042-2042
Publication Date(Web):2013/01/25
DOI:10.1039/C3CC38687B
Based on an azo–thiol redox reaction, a non-emissive dinuclear iridium(III) complex showed off–on phosphorescent response toward thiols and was developed to image thiol levels in living cells.
Co-reporter:Guo-Liang Liao, Xiang Chen, Liang-Nian Ji and Hui Chao
Chemical Communications 2012 - vol. 48(Issue 87) pp:NaN10783-10783
Publication Date(Web):2012/09/13
DOI:10.1039/C2CC36039J
A novel ruthenium(II) complex with specific luminescent selectivity towards hybrid G-quadruplex DNA was developed that can easily be distinguished by the naked eye without further treatment.
Co-reporter:Huaiyi Huang, Pingyu Zhang, Yu Chen, Kangqiang Qiu, Chengzhi Jin, Liangnian Ji and Hui Chao
Dalton Transactions 2016 - vol. 45(Issue 33) pp:NaN13145-13145
Publication Date(Web):2016/05/25
DOI:10.1039/C6DT01270A
DNA binding and DNA transcription inhibition is regarded as a promising strategy for cancer chemotherapy. Herein, chloro terpyridyl Ru(II) complexes, [Ru(tpy)(N^N)Cl]+ (Ru1, N^N = 2,2′-bipyridine; Ru2, N^N = 3-(pyrazin-2-yl)-as-triazino[5,6-f]acenaphthylene; Ru3, N^N = 3-(pyrazin-2-yl)-as-triazino[5,6-f]phenanthrene; Ru4, N^N = 3-(pyrazin-2-yl)-as-triazino[5,6-f]pyrene) were prepared as DNA intercalative and covalent binding anticancer agents. The chloro ligand hydrolysis slowly and the octanol and water partition coefficient of Ru2–Ru4 were between 0.6 and 1.2. MALDI-TOF mass, DNA gel electrophoresis confirmed covalent and intercalative DNA binding modes of Ru2–Ru4, while Ru1 can only bind DNA covalently. As a result, Ru2–Ru4 exhibited stronger DNA transcription inhibition activity, higher cell uptake efficiency and better anticancer activity than Ru1. Ru4 was the most toxic complex toward all cancer cells which inhibited DNA replication and transcription. AO/EB, Annexin V/PI, nuclear staining, JC-1 assays further confirmed that Ru2–Ru4 induced cancer cell death by an apoptosis mechanism.
Co-reporter:Kangqiang Qiu, Bole Yu, Huaiyi Huang, Pingyu Zhang, Liangnian Ji and Hui Chao
Dalton Transactions 2015 - vol. 44(Issue 15) pp:NaN7065-7065
Publication Date(Web):2015/03/09
DOI:10.1039/C5DT00117J
To prolong the observation time, increase the penetration depth and decrease self-absorption and phototoxicity, two-photon luminescent vectors have emerged as promising tools for tracking gene delivery in living cells. Herein, we report four new tetranuclear Ru(II) complexes based on oligo-oxyethylene and polybenzimidazole as one- and two- photon luminescent tracking non-viral gene vectors. In such a molecular design, the oligo-oxyethylene, polybenzimidazole and Ru(II) polypyridyl complexes were expected to render the vectors with increased cell permeability, biocompatibility, proton buffering capacity and one- and two-photon luminescence. Corresponding DNA interaction studies showed that the ability of the complexes to condense DNA decreased with increasing oligo-oxyethylene lengths. Additionally, all complexes protected DNA. The complexes were investigated as one- and two-photon tracking non-viral gene vectors in living cells and showed proper cellular uptake, good luciferase expression and low cytotoxicity.
Co-reporter:Jin-Quan Wang, Jun-Feng Kou, Zi-Zhuo Zhao, Kang-Qiang Qiu and Hui Chao
Inorganic Chemistry Frontiers 2017 - vol. 4(Issue 6) pp:NaN1012-1012
Publication Date(Web):2017/04/17
DOI:10.1039/C7QI00149E
Metastasis is a major health threat for most cancer patients, thus anti-metastasis treatments that reduce cell migration and invasion are critical for cancer treatment. In this study, four anthraquinone-bridged diruthenium(II) complexes, [(bpy)2Ru(L)Ru(bpy)2]4+ (Ru1, L = 1,4-bis(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)anthracene-9,10-dione; Ru2, L = 1,5-bis(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)anthracene-9,10-dione; Ru3, L = 2,6-bis(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)anthracene-9,10-dione; and Ru4, L = 2,7-bis(1H-imidazo[4,5-f][1,10]phenanthrolin-2-yl)anthracene-9,10-dione) were synthesized and characterized. These Ru(II) complexes exhibited multi-targeted anti-metastatic properties against human hepatocarcinoma MHCC97-H cells that included the inhibition of migration and invasion. Further investigation of the intracellular mechanisms revealed that Ru(II) complexes suppressed the phosphorylation of ERK and AKT. Moreover, significant reduction of the extracellular and intracellular expression of the metastatic regulatory proteins MMP-2 and MMP-9 was also observed after Ru1–Ru4 treatment. In addition, these Ru(II) complexes negatively modulate the actin cytoskeleton by inhibiting Cdc42 protein expression, arresting the cells in the G2/M phase. The results indicate that these ruthenium(II) complexes have potential as drug candidates for anti-metastatic therapies.
Co-reporter:Leli Zeng, Shi Kuang, Guanying Li, Chengzhi Jin, Liangnian Ji and Hui Chao
Chemical Communications 2017 - vol. 53(Issue 12) pp:NaN1980-1980
Publication Date(Web):2017/01/19
DOI:10.1039/C6CC10330H
A glutathione (GSH)-activatable ruthenium(II)-azo photosensitizer was prepared. The complex had low toxicity towards cells under dark conditions. It exhibited excellent phototoxicity under two-photon excitation (810 nm) and thus was developed as a two-photon photodynamic anticancer agent for cancer therapy.
Co-reporter:Jiangping Liu, Chengzhi Jin, Bo Yuan, Xingguo Liu, Yu Chen, Liangnian Ji and Hui Chao
Chemical Communications 2017 - vol. 53(Issue 12) pp:NaN2055-2055
Publication Date(Web):2017/01/18
DOI:10.1039/C6CC10015E
Herein a series of mitochondria-targeted AIE (aggregation-induced emission)-active Ir(III) complexes were designed to selectively exert one-/two-photon photodynamic activities in mitochondria to address the issues which current PDT are confronted with (i.e., shallow penetration depth of routinely used irradiation; systematic toxicity associated with effective drug concentration; concentration-quenched photodynamic activity at the target, etc.).