Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ting Meng is active.

Publication


Featured researches published by Ting Meng.


Journal of Medicinal Chemistry | 2015

Stabilization of G-quadruplex DNA, inhibition of telomerase activity, and tumor cell apoptosis by organoplatinum(II) complexes with oxoisoaporphine.

Zhen-Feng Chen; Qi-Pin Qin; Jiao-Lan Qin; Yan-Cheng Liu; Ke-Bin Huang; Yu-Lan Li; Ting Meng; Guo-Hai Zhang; Yan Peng; Xu-Jian Luo; Hong Liang

Two G-quadruplex ligands [Pt(L(a))(DMSO)Cl] (Pt1) and [Pt(L(b))(DMSO)Cl] (Pt2) have been synthesized and fully characterized. The two complexes are more selective for SK-OV-3/DDP tumor cells versus normal cells (HL-7702). It was found that both Pt1 and Pt2 could be a telomerase inhibitor targeting G-quadruplex DNA. This is the first report demonstrating that telomeric, c-myc, and bcl-2 G-quadruplexes and caspase-3/9 preferred to bind with Pt2 rather than Pt1, which also can induce senescence and apoptosis. The different biological behavior of Pt1 and Pt2 may correlate with the presence of a 6-hydroxyl group in L(b). Importantly, Pt1 and Pt2 exhibited higher safety in vivo and more effective inhibitory effects on tumor growth in the HCT-8 and NCI-H460 xenograft mouse model, compared with cisplatin. Taken together, these mechanistic insights indicate that both Pt1 and Pt2 display low toxicity and could be novel anticancer drug candidates.


European Journal of Medicinal Chemistry | 2016

Stabilization of c-myc G-Quadruplex DNA, inhibition of telomerase activity, disruption of mitochondrial functions and tumor cell apoptosis by platinum(II) complex with 9-amino-oxoisoaporphine.

Jiao-Lan Qin; Qi-Pin Qin; Zu-Zhuang Wei; Yan-Cheng Yu; Ting Meng; Chen-Xuan Wu; Yue-Lan Liang; Hong Liang; Zhen-Feng Chen

[Pd(L)(DMSO)Cl2] (1) and [Pt(L)(DMSO)Cl2] (2) with 9-amino-oxoisoaporphine (L), were synthesized and characterized. 1 and 2 are more selectively cytotoxic to Hep-G2 cells versus normal liver cells (HL-7702). Various experiments showed that 2 acted as telomerase inhibitors targeting G4-DNA and triggered cell apoptosis by interacting with c-myc G4-DNA. Furthermore, 2 significantly induced cell cycle arrest at both G2/M and S phase, which leading to the down-regulation of cdc25 A, cyclin D, cyclin B, cyclin A and CDK2 and the up-regulation of p53, p27, p21,chk1 and chk2. In addition, 2 also caused mitochondrial dysfunction. Taken together, we found that 2 exerted its cytotoxic activity mainly via inhibiting telomerase by interaction with c-myc G4-DNA and disruption of mitochondrial function.


MedChemComm | 2015

Cytotoxicity, DNA binding and cell apoptosis induction of a zinc(II) complex of HBrQ

Hai-Rong Zhang; Yan-Cheng Liu; Ting Meng; Qi-Pin Qin; Shang-Feng Tang; Zhen-Feng Chen; Bi-Qun Zou; You-Nian Liu; Hong Liang

A new zinc(II) complex (1) of 5-bromo-8-hydroxyquinoline (HBrQ) was prepared and structurally characterized using IR, ESI-MS, elemental analysis, 1H and 13C NMR, as well as single crystal X-ray diffraction analysis. The DNA binding study on complex 1, which was performed using UV-vis, fluorescence and circular dichroism (CD) spectral analyses, suggested that complex 1 interacts with ct-DNA mainly via an intercalative binding mode. The in vitro cytotoxicity of complex 1, compared with Zn(OAc)2·H2O, HBrQ and cisplatin, was screened against a series of tumor cell lines as well as the normal liver cell line HL-7702 using the MTT assay. Complex 1 showed much higher cytotoxicity than Zn(OAc)2·H2O and HBrQ against most of the tumor cell lines, in which BEL-7404 was the most sensitive tumor cell line towards 1, with an IC50 value of 8.69 ± 0.04 μM. Complex 1 was found to greatly induce cell cycle arrest in the BEL-7404 cells at the G2 phase, and consequently to induce cell apoptosis in a dose-dependent mode, which is suggested by the cell apoptosis analysis via the Hoechst 33258 and AO/EB staining assays. Targeting the mitochondria pathway due to the redox activity of Zn, the apoptotic mechanism in the BEL-7404 cells treated by 1 was investigated using reactive oxygen species (ROS) detection, intracellular calcium concentration measurement and caspase-9/3 activity assay, which showed that the cell apoptosis induced by 1 was closely related to the loss of mitochondrial membrane potential, ROS production and enhancement of intracellular [Ca2+], which trigger caspase-9/3 activation via the mitochondrial dysfunction pathway.


Scientific Reports | 2016

Preparation of 6/8/11-Amino/Chloro-Oxoisoaporphine and Group-10 Metal Complexes and Evaluation of Their in Vitro and in Vivo Antitumor Activity

Qi-Pin Qin; Jiao-Lan Qin; Ting Meng; Gui-Ai Yang; Zu-Zhuang Wei; Yan-Cheng Liu; Hong Liang; Zhen-Feng Chen

A series of group-10 metal complexes 1–14 of oxoisoaporphine derivatives were designed and synthesized. 1–14 were more selectively cytotoxic to Hep-G2 cells comparing with normal liver cells. In vitro cytotoxicity results showed that complexes 1–6, 7, 8, 10 and 11, especially 3, were telomerase inhibitors targeting c-myc, telomeric, and bcl-2 G4s and triggered cell senescence and apoptosis; they also caused telomere/DNA damage and S phase arrest. In addition, 1–6 also caused mitochondrial dysfunction. Notably, 3 with 6-amino substituted ligand La exhibited less side effects than 6 with 8-amino substituted ligand Lb and cisplatin, but similar tumor growth inhibition efficacy in BEL-7402 xenograft model. Complex 3 has the potential to be developed as an effective anticancer agent.


MedChemComm | 2016

Evaluation of the effect of iodine substitution of 8-hydroxyquinoline on its platinum(II) complex: cytotoxicity, cell apoptosis and telomerase inhibition

Ting Meng; Shang-Feng Tang; Qi-Pin Qin; Yue-Lan Liang; Chen-Xuan Wu; Chen-Yu Wang; Hou-Tian Yan; Jia-Xin Dong; Yan-Cheng Liu

Two platinum(II) complexes, [PtCl(Q)(DMSO)] (1) and [PtCl(IQ)(DMSO)] (2), bearing 8-hydroxyquinoline (H-Q) and 5,7-diiodo-8-hydroxyquinoline (H-IQ) as the bioactive ligand, respectively, were synthesized and structurally characterized. By MTT assay, complex 2 bearing the IQ ligand showed significantly higher growth inhibition than complex 1 against all the five typical tumor cell lines in the test, but showed no more cytotoxicity against the normal liver cell line HL-7702, suggesting the much better cytotoxic selectivity of 2 than that of 1. In addition, the HepG2 cell line was found to be the most sensitive towards both complexes. Aiming at the HepG2 cell line, both complexes arrested the cell cycle of HepG2 cells in the S phase, as examined by flow cytometry, in which complex 2 showed higher S-phase arrest than 1. This was supported by the down-regulation of cdc25 A, cyclin B, cyclin A, and CDK2 and the up-regulation of p53, p27 and p21 based on western blot assay. Complex 2 also acted as a more effective telomerase inhibitor than 1 by interacting with telomeric/c-myc G-quadruplexes and triggering cell senescence and cell apoptosis. Furthermore, both complexes caused mitochondrial dysfunction, suggesting a potential mitochondrion-mediated apoptotic pathway induced by each complex. From the platinum uptake assay, complex 2 exhibited obvious priority on the cell uptake effect than 1, which should be undoubtedly correlated with the key roles of the 5- and 7-iodo-substituted groups in the IQ ligand of 2. This may well explain the better cytotoxicity and the more significant antitumor mechanism of 2 throughout the study. This work further demonstrated that rational halogen substitution on selected ligands would be greatly beneficial to achieve more promising metal-based antitumor agents.


MedChemComm | 2016

Cobalt(II) 8-hydroxyquinoline complexes: structure, cytotoxicity and action mechanism

Hai-Rong Zhang; Ke-Bin Huang; Zhen-Feng Chen; Yan-Cheng Liu; You-Nian Liu; Ting Meng; Qi-Pin Qin; Bi-Qun Zou; Hong Liang

Three cobalt(II) complexes, [Co(NOQ)2(C5H5N)2]C2H5OH (1), [Co(BrQ)2(C5H5N)2] (2) and [Co(NQ)2(C5H5N)2] (3) (NOQ = 5-nitro-8-hydroxyquinoline, BrQ = 5-bromo-8-hydroxyquinoline, NHQ = 2-amino-8-hydroxyquinoline), were synthesized and characterized. Their in vitro cytotoxicity against T-24, BEL-7404, HepG2, HeLa, MGC-803, SKOV-3, HL-7702 and WI-38 cell lines was evaluated. Complexes 1–3 exhibited the highest proliferation inhibition activity against the T-24 tumor cell line with IC50 values in the range between 7.00 and 16.70 μM. They also displayed selectivity towards T-24 tumor cell lines compared to the normal liver cell line HL-7702 and human fetal lung fibroblast WI-38 cell line. The action mechanism of complex 1 was investigated. It caused apoptotic death of the T-24 cells via G1 cell cycle arrest. Further investigation revealed that complex 1 induced overproduction of reactive oxygen species, which led to mitochondrion-mediated apoptosis. DNA binding studies suggested that intercalation might be the most probable binding mode of the Co(II) complexes with ct-DNA.


New Journal of Chemistry | 2016

Studies on the structures, cytotoxicity and apoptosis mechanism of 8-hydroxylquinoline rhodium(III) complexes in T-24 cells

Hai-Rong Zhang; Yan-Cheng Liu; Zhen-Feng Chen; Ting Meng; Bi-Qun Zou; You-Nian Liu; Hong Liang

Two rhodium(III) complexes (Rh(OQ)3 (1) and Rh(BrQ)2(CH3OH)Cl (2), HOQ = 8-hydroxyquinoline, HBrQ = 5-bromo-8-hydroxyquinoline) of 8-hydroxylquinoline were synthesized and characterized. By MTT assay, the in vitro cytotoxicity of complexes 1 and 2, compared with HOQ, HBrQ and cisplatin, was evaluated towards a series of tumor cell lines as well as the normal liver cell line HL-7702. Complexes 1 and 2 showed higher cytotoxicity against the tested tumor cell lines than the corresponding ligands, among which T-24 was the most sensitive cell line for complexes 1 and 2 (IC50 = 13.42 μM for 1, 18.91 μM for 2). Compared with cisplatin, complex 1 exhibited higher cytotoxicity against T-24 cells but lower cytotoxicity against HL-7702(IC50 = 15.93 μM). Considering the better cytotoxicity of complex 1 than complex 2 against T-24 cells, the underlying anticancer molecular mechanisms were also investigated. DNA interaction studies revealed that complex 1 interacted with ct-DNA mainly via an intercalative binding mode. Further investigation of intracellular mechanisms revealed that complex 1 caused G2 phase cell cycle arrest and induced T-24 cell apoptosis in a dose-dependent mode. Targeting the mitochondrial pathway, the apoptotic mechanism in T-24 cells treated with 1 was studied by ROS detection, intracellular Ca2+ concentration measurements and caspase-9/3 activity assay, which suggested that complex 1 induced T-24 cell apoptosis by the disruption of mitochondrial-related mechanisms.


Chemical & Pharmaceutical Bulletin | 2016

Synthesis, structure characterization and antitumor activity study of a new iron(III) complex of 5-nitro-8-hydroxylquinoline (HNOQ)

Hai-Rong Zhang; Ting Meng; Yan-Cheng Liu; Qi-Pin Qin; Zhen-Feng Chen; You-Nian Liu; Hong Liang

A new iron(III) complex (1) of 5-nitro-8-hydroxylquinoline (HNOQ) was synthesized and structurally characterized in its solid state and solution state by IR, UV-Vis, electrospray ionization (ESI)-MS, elemental analysis, conductivity and X-ray single crystal diffraction analysis. The DNA binding study suggested that complex 1 interacted with calf thymus (ct)-DNA mainly via an intercalative binding mode. By 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, the in vitro cytotoxicity of complex 1, comparing with HNOQ and cisplatin, was screened towards a series of tumor cell lines as well as the normal liver cell line HL-7702. Complex 1 showed higher cytotoxicity towards the tested tumor cell lines but lower cytotoxicity towards HL-7702 than HNOQ, in which the T-24 was the most sensitive cell line for 1. Complex 1 caused G2 phase cell cycle arrest and induced cell apoptosis in T-24 cells in a dose-dependent mode, evidenced by changes in cell morphology. Targeting the mitochondrial pathway due to the redox potential of Fe(III)/Fe(II), the apoptotic mechanism in T-24 cells treated by 1 was investigated by reactive oxygen species (ROS) detection, intracellular [Ca(2+)] measurement and caspase-9 and caspase-3 activity assay. It suggested that complex 1 induced cell apoptosis by triggering the caspase-9 and caspase-3 activation via a mitochondrion-mediated pathway.


Oncotarget | 2017

Chiral platinum (II)-4-(2,3-dihydroxypropyl)- formamide oxo-aporphine (FOA) complexes promote tumor cells apoptosis by directly targeting G-quadruplex DNA in vitro and in vivo

Qi-Pin Qin; Jiao-Lan Qin; Ming Chen; Yu-Lan Li; Ting Meng; Jie Zhou; Hong Liang; Zhen-Feng Chen

Three platinum(II) complexes, 4 (LC-004), 5 (LC-005), and 6 (LC-006), with the chiral FOA ligands R/S-(±)-FOA (1), R-(+)-FOA (2) and S-(–)-FOA (3), respectively, were synthesized and characterized. As potential anti-tumor agents, these complexes show higher cytotoxicity to BEL-7404 cells than the HL-7702 normal cells. They are potential telomerase inhibitors that target c-myc and human telomeric G-quadruplex DNA. Compared to complexes 4 and 5, 6 exhibited higher binding affinities towards telomeric, c-myc G-quadruplex DNA and caspase-3/9, thereby inducing senescence and apoptosis to a greater extent in tumor cells. Moreover, our in vivo studies showed that complex 6 can effectively inhibit tumor growth in the BEL-7404 and BEL-7402 xenograft mouse models and is less toxic than 5-fluorouracil and cisplatin. The effective inhibition of tumor growth is attributed to its interactions with 53BP1, TRF1, c-myc, TRF2, and hTERT. Thus, complex 6 can serve as a novel lead compound and a potential drug candidate for anticancer chemotherapy.Three platinum(II) complexes, 4 (LC-004), 5 (LC-005), and 6 (LC-006), with the chiral FOA ligands R/S-(±)-FOA (1), R-(+)-FOA (2) and S-(-)-FOA (3), respectively, were synthesized and characterized. As potential anti-tumor agents, these complexes show higher cytotoxicity to BEL-7404 cells than the HL-7702 normal cells. They are potential telomerase inhibitors that target c-myc and human telomeric G-quadruplex DNA. Compared to complexes 4 and 5, 6 exhibited higher binding affinities towards telomeric, c-myc G-quadruplex DNA and caspase-3/9, thereby inducing senescence and apoptosis to a greater extent in tumor cells. Moreover, our in vivo studies showed that complex 6 can effectively inhibit tumor growth in the BEL-7404 and BEL-7402 xenograft mouse models and is less toxic than 5-fluorouracil and cisplatin. The effective inhibition of tumor growth is attributed to its interactions with 53BP1, TRF1, c-myc, TRF2, and hTERT. Thus, complex 6 can serve as a novel lead compound and a potential drug candidate for anticancer chemotherapy.


Oncotarget | 2017

Facile total synthesis of lysicamine and the anticancer activities of the Ru II , Rh III , Mn II and Zn II complexes of lysicamine

Jiao-Lan Qin; Ting Meng; Zhen-Feng Chen; Xiao-Li Xie; Qi-Pin Qin; Xiao-Ju He; Ke-Bin Huang; Hong Liang

Lysicamine is a natural oxoaporphine alkaloid, which isolated from traditional Chinese medicine (TCM) herbs and has been shown to possess cytotoxicity to hepatocarcinoma cell lines. Reports on its antitumor activity are scarce because lysicamine occurs in plants at a low content. In this work, we demonstrate a facile concise total synthesis of lysicamine from simple raw materials under mild reaction conditions, and the preparation of the Ru(II), Rh(III), Mn(II) and Zn(II) complexes 1–4 of lysicamine (LY). All the compounds were fully characterized by elemental analysis, IR, ESI-MS, 1H and 13C NMR, as well as single-crystal X-ray diffraction analysis. Compared with the free ligand LY, complexes 2 and 3 exhibited superior in vitro cytotoxicity against HepG2 and NCI-H460. Mechanistic studies indicated that 2 and 3 blocked the cell cycle in the S phase by decreasing of cyclins A2/B1/D1/E1, CDK 2/6, and PCNA levels and increasing levels of p21, p27, p53 and CDC25A proteins. In addition, 2 and 3 induced cell apoptosis via both the caspase-dependent mitochondrial pathway and the death receptor pathway. in vivo study showed that 2 inhibited HepG2 tumor growth at 1/3 maximum tolerated dose (MTD) and had a better safety profile than cisplatin.

Collaboration


Dive into the Ting Meng's collaboration.

Top Co-Authors

Avatar

Hong Liang

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Qi-Pin Qin

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Yan-Cheng Liu

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Zhen-Feng Chen

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Jiao-Lan Qin

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Bi-Qun Zou

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Hai-Rong Zhang

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

You-Nian Liu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Zu-Zhuang Wei

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Ke-Bin Huang

Guangxi Normal University

View shared research outputs
Researchain Logo
Decentralizing Knowledge