Xue-Yi Le
South China Agricultural University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Xue-Yi Le.
Journal of Inorganic Biochemistry | 2015
Xia-Bing Fu; Jia-Jia Zhang; Dan-Dan Liu; Qian Gan; Hong-Wei Gao; Zong-Wan Mao; Xue-Yi Le
Two new Cu(II)-dipeptide complexes of 2-(4-thiazolyl)benzimidazole, [Cu(Gly-Gly)(TBZ)(Cl)]·4H2O (1) and [Cu(Gly-l-Leu)(TBZ)(Cl)]·H2O (2) (Gly-Gly=glycyl-glycine anion, Gly-l-Leu=glycyl-l-leucine anion and TBZ=2-(4-thiazolyl)benzimidazole) have been synthesized and characterized by elemental analyses, molar conductance measurements and spectroscopy methods (IR, UV-visible, electrospray ionization mass spectra (ESI-MS) and EPR). The DNA binding and cleavage properties of the complexes monitored by multi-spectroscopic techniques (UV absorption, fluorescence and circular dichroism), viscosity determination and agarose gel electrophoresis indicated that the complexes bound to calf thymus (CT)-DNA via a partial intercalative mode with considerable intrinsic binding constants (Kb=1.64×10(5)M(-1) for 1 and 2.59×10(5)M(-1) for 2), and cleaved pBR322 DNA efficiently in the mediation of ascorbic acid (AA), probably via an oxidative damage mechanism induced by OH. The antioxidant activities of the complexes have been evaluated by means of modified nitroblue tetrazolium (NBT) photoreduction and cellular antioxidant activity (CAA) assays using HepG2 cells as a model, and it was found that IC50 values of 1 and 2 for dismutation of O2(-) were 0.172 and 0.247μM, respectively, and the CAA50 values were 10.57 and 10.74μM. In addition, the complexes were subjected to in vitro cytotoxicity against three human carcinoma cell lines (HeLa, A549 and HepG2), which revealed that the complexes exhibited effective cytotoxicity (IC50 values varying from 33.17 to 100μM) and selective inhibition toward HeLa cell lines. These findings indicate that the complexes have the potential to act as effective metallopeptide chemotherapeutic agents.
Journal of Inorganic Biochemistry | 2013
Wei Hu; Suwen Deng; Jianyin Huang; Yanmei Lu; Xue-Yi Le; Wenxu Zheng
The intercalative interactions of small molecules with DNA are important in a variety of biological processes including mutagenesis, carcinogenesis, and chemotherapy. A comprehensive research protocol including experiments and calculations was employed to investigate the intercalative interaction between metallointercalator copper(II) complex and DNA. The intercalative binding mode has been validated by UV spectra, fluorescence spectra, CD spectra and viscosity measurements. The classical molecular dynamics simulation was carried out to investigate the intercalative interaction between asymmetric copper(II) complex and DNA. An analytical method was proposed to simulate the dynamically changing absorption spectra of intercalator/DNA system. According to the established model, the changing process of the electronic absorption spectra for intercalator/DNA system can be predicted accurately. A rational explanation for the change law of absorption spectra has been proposed. Moreover, the analyses of the frontier orbital reveal that the red shift of the absorption spectra is due to the increase of π orbital energy caused by the coupling of the π orbital of the intercalated ligand with the π orbital of DNA. This cause of red shift of spectra is completely different from the previous inference. All these insights are of crucial importance for correctly analyzing the absorption spectra of intercalative interaction, as well as for explaining the macroscopic phenomena observed in experiments at the molecular level.
RSC Advances | 2016
Qian Gan; Chun-Lian Zhang; Bing-Feng Wang; Ya-Hong Xiong; Yinlian Fu; Zong-Wan Mao; Xue-Yi Le
Two novel mononuclear mixed ligand copper(II) complexes, [Cu(Gly-L-val)(HPB)(H2O)]·ClO4·1.5H2O (1) and [Cu(Gly-L-val)(PBT)(H2O)]·ClO4 (2) (Gly-L-val = glycyl-L-valine, HPB = 2-(2′-pyridyl)benzimidazole, PBT = 2-(2′-pyridyl)benzothiazole), have been synthesized and characterized using various analytical and spectroscopic methods. The interactions of the complexes with DNA have been explored by viscometry, thermal denaturation, cyclic voltammetry (CV), agarose gel electrophoresis and spectroscopic means (UV absorption, circular dichroism (CD) and fluorimetry), as well as molecular docking techniques. These studies confirmed the mode of the complexes bound to calf thymus DNA (CT-DNA) through insertion with certain affinity (Kb = 3.211 × 105 M−1 for 1 and 4.734 × 104 M−1 for 2). In the fluorimetric experiments of thermodynamics (KSV = 1.145 × 104 M−1 for 1 and 2.634 × 103 M−1 for 2), the changes in enthalpy (ΔH > 0), entropy (ΔS > 0) and Gibbs free energy (ΔG < 0) in the interactions between the complexes with DNA suggested that the process occurred spontaneously through hydrophobic interactions. The complexes displayed oxidative cleavage of pBR322 plasmid DNA in the presence of ascorbic acid, probably induced by ˙OH as a reactive oxygen species. Furthermore, the molecular docking technique was applied to ascertain the mode of action for the complexes towards DNA. Moreover, superoxide dismutase (SOD) activity studies were performed using the photoreduction of nitroblue tetrazolium (NBT) under a non-enzymatic system and the antioxidant activities of 1 and 2 determined with IC50 values of 0.337 and 0.146 μM, respectively. The cytotoxicity of the Cu(II) complexes against A549, HeLa, PC-3 tumor cell lines and NIH3T3 (non-tumor cell line) was studied by an MTT assay and it was found that 1 exhibited better cytotoxicity against A549 and PC-3 than 2 and the widely used drug cisplatin.
Journal of Fluorescence | 2016
Qian Gan; Xia-Bing Fu; Weijiang Chen; Ya-Hong Xiong; Yinlian Fu; Shi Chen; Xue-Yi Le
A new mixed ligand copper(II)-dipeptide complex with 2-(2′-pyridyl)benzothiazole (pbt), [Cu(Gly-L-leu)(pbt)(H2O)]·ClO4 (Gly-L-leuxa0=xa0Glycyl-L-leucine anion) was synthesized and characterized by various physico-chemical means. The DNA binding and cleavage properties of the complex investigated by viscosity, agarose gel electrophoresis and multi-spectroscopic techniques (UV, circular dichroism (CD) and fluorescence) showed that the complex was bound to CT-DNA through intercalation mode with moderate binding constant (Kbxa0=xa03.132xa0×xa0104xa0M−1), and cleaved pBR322 DNA efficiently (~ 5xa0μM) in the presence of Vc, probably via an oxidative mechanism induced by •OH. Additionally, the interaction of the complex with human serum albumin (HSA) was explored by UV-visible, CD, fluorescence, synchronous fluorescence and 3D fluorescence spectroscopy. The complex exhibits desired affinity to HSA through hydrophobic interaction. Moreover, the cytotoxicity of the complex against three human carcinoma cell lines (HeLa, HepG2 and A549) was evaluated by MTT assay, which showed that the complex had effective cytotoxicity and higher inhibition toward A549 cell lines with IC50 of 38.0xa0±xa03.2xa0μM.
Journal of Fluorescence | 2017
Qian Gan; Yong-Yu Qi; Ya-Hong Xiong; Yinlian Fu; Xue-Yi Le
Two new mononuclear mixed ligand copper(II) complexes [Cu(PBO)(Gly-gly)(H2O)]·ClO4·1.5H2O (1) and [Cu(PBO)(Gly-L-leu)(H2O)]·ClO4 (2) (PBO is 2-(2′-pyridyl)benzoxazole, Gly-gly and Gly-L-leu are Glycyl-glycine anion and Glycyl-L-leucine anion, respectively), have been prepared and characterized by various analytical and spectral techniques. The interactions of the complexes with DNA were investigated using multi-spectroscopic methods (absorption, emission, circular dichroism), viscometry and electrochemical titration as well as molecular docking technique. The results indicated that 1 and 2 are bound to calf thymus DNA (CT-DNA) through an intercalative mode. The thermodynamic analyses revealed that the reactions between the Cu(II) complexes with DNA are spontaneous with negative Gibbs free energy (ΔG). The positive changes of enthalpy (ΔH) and entropy (ΔS) suggested that the binding processes are dominated by hydrophobic interaction accompanying with endothermic. Also, the complexes exhibited efficient oxidative cleavage of pBR322 plasmid DNA in the presence of ascorbic acid, probably induced by •OH as reactive oxygen species. In addition, 1 and 2 displayed excellent antioxidant activities with the IC50 values of 0.112 and 0.191xa0μM, respectively, using the mean of nitroblue tetrazolium (NBT) photochemical reduction under a nonenzymatic condition. Moreover, the complexes were screened for their in vitro cytotoxicity against three human carcinoma cell lines (HeLa, PC-3 and A549), in which 2 owns higher cytotoxicity, which was consistent with DNA binding and cleavage ability order of the complexes. This results showed the in vitro biochemical potentials of the Cu(II)-dipeptide complexes with aromatic heterocyclic, viz. effective metallopeptide-nucleases, SOD mimics and non-platinum chemotherapeutic metallopharmaceuticals and their structure-activity relationship, which may contribute to the rational molecular design of new metallopeptide based chemotherapeutic agents.
New Journal of Chemistry | 2017
Fang Shen; Ya-Xian Liu; Shu-Min Li; Chi-Kun Jiang; Bing-Feng Wang; Ya-Hong Xiong; Zong-Wan Mao; Xue-Yi Le
Two new mixed-ligand copper(II) complexes, [Cu(PyTA)(L-Phe)Cl]·2H2O (1), [Cu(PyTA)(L-Met)Cl]·0.5H2O (2) (where PyTA = 2,4-diamino-6-(2′-pyridyl)-1,3,5-triazine, L-Phe = L-phenylalanine, L-Met = L-methionine), were synthesized and characterized by elemental analysis, IR, UV-vis, molar conductivity, ESI-MS, and X-ray crystal diffraction. The copper centers of complexes 1 and 2 are in slightly distorted square pyramidal environments, and fluorescence quenching experiments and calorimetry showed that the complexes bound to human serum albumin (HSA) with high affinities (binding constants KD = 3.71 × 106 L mol−1 for 1 and 1.99 × 106 L mol−1 for 2, 1 > 2). UV-vis and circular dichroism (CD) spectroscopic investigations revealed that the complexes induced the alteration of hydrophobic environments and the decrease in the α-helix level of HSA during binding processes. Synchronous fluorescence spectra demonstrated that the binding sites of the complexes on HSA were close to the Trp-214 residue in the IIA subdomain, which was further verified by site marker competitive experiments and the molecular docking method. In vitro cytotoxicity experiments showed that the effective HSA-binding interactions enhanced the cytotoxicity of the complexes. Both 1 and 2 displayed strong anticancer activities against the Eca-109 cell line and induced cell death through the apoptosis pathway.
Transition Metal Chemistry | 2018
Ya-Xian Liu; Hui-Wen Mo; Zhen-Yu Lv; Fang Shen; Chun-Lian Zhang; Yong-Yu Qi; Zong-Wan Mao; Xue-Yi Le
A copper complex [Cu(HPBM)(l-Phe)(H2O)]·ClO4(1) (HPBMxa0=xa05-methyl-2-(2′-pyridyl)benzimidazole, l-Phexa0=xa0l-phenylalanine anion) was synthesized and characterized by elemental analysis, IR, ESI–MS, HR–ESI–MS, ESR spectroscopy, and by X-ray single-crystal analysis. The binding constant of the complex with calf thymus DNA (CT-DNA) was determined as 7.38 (±xa00.57)xa0×xa0104xa0M−1. Further studies indicated that the complex interacts with CT-DNA through minor groove binding. The in vitro cytotoxic activities of both the free proligand and the complex against Eca-109, HeLa and A549 cancer cells and normal LO2 cells were evaluated by the MTT method. The IC50 values range from 5.7xa0±xa00.1 to 8.3xa0±xa00.6xa0µM. Free HPBM displays no cytotoxic activity against the selected cancer cells, with IC50 values more than 100xa0µM. Double staining analysis showed that the complex can induce apoptosis in Eca-109 cells. Comet assays demonstrated that the complex can damage DNA and cause apoptosis. The complex also induces an increase in intracellular reactive oxygen species and a reduction in mitochondrial membrane potential. The complex can also increase the intracellular Ca2+ level and induce release of cytochrome c. The cell cycle arrest was investigated by flow cytometry. The results demonstrate that the complex induces apoptosis in Eca-109 cells through DNA-binding and ROS-mediated mitochondrial dysfunctional pathways.
Transition Metal Chemistry | 2018
Chun-Lian Zhang; Xue-Mei Zhang; Wei Liu; Shi Chen; Xue-Yi Le
A water-soluble Cu(II) complex, [Cu(pzta)(L-Ser)(ClO4)]·1.5H2O (pztaxa0=xa06-(pyrazin-2-yl)-1,3,5-triazine-2,4-diamine; L-Serxa0=xa0L-serinate), was synthesized and characterized by analytical and spectral techniques. In addition, the molecular structure of the complex was confirmed by single-crystal X-ray diffraction, revealing that the central Cu(II) atom was located in a six-coordinate distorted octahedral geometry. Multi-spectroscopic methods, viscosity measurements and thermal denaturation experiments revealed that the complex binds to DNA with apparent binding constant of 2.93xa0×xa0103xa0M−1 through a groove binding mode. The positive values of ΔH and ΔS obtained from isothermal titration calorimetry experiments indicated that hydrophobic interactions play an important role in the formation of the complex–DNA adduct. Molecular docking studies were carried out to better understand the binding mode of the complex with DNA.
Materials Science and Engineering: C | 2018
Chun-Lian Zhang; Ya-Xian Liu; Xue-Mei Zhang; Shi Chen; Fang Shen; Ya-Hong Xiong; Wei Liu; Zong-Wan Mao; Xue-Yi Le
Two water-soluble copper(II) complexes of 6-(pyrazin-2-yl)-1,3,5-triazine-2,4-diamine (pzta) and amino acids, [Cu(pzta)(L-ArgH)(H2O)](ClO4)2 (1) and [Cu(pzta)(L-Met)(H2O)]ClO4·3H2O (2) (L-ArgH: protonated L-Argininate; L-Met: L-Methioninate), were synthesized and characterized. The determined X-ray crystallographic structures of 1 and 2 exhibited distorted square-pyramidal coordination geometries. Their binding properties toward calf thymus DNA (CT-DNA) and human serum protein (HSA) were measured by spectroscopic (UV-Vis, fluorescence, circular dichroism (CD)), calorimetric (isothermal titration calorimetry (ITC)) and molecular docking technology. DNA binding experiments showed that the complexes bound to DNA through a groove binding mode, the positive ΔH and ΔS values indicated that the hydrophobic interaction was the main force in the binding between the complexes and DNA. Besides, the complexes caused the fluorescence quenching of HSA through a static quenching procedure, changed the secondary structure and microenvironment of the Trp-214 residue, and preferably bound to subdomain IIA of HSA driven by hydrophobic and hydrogen-bond interactions. These results were further verified by the molecular docking technology. Furthermore, the in vitro cytotoxicities of the complexes against three human carcinoma cell lines (A549, PC-3 and HeLa) were evaluated, which confirmed that the complexation improved the anticancer activity of the pzta ligand significantly.
International Journal of Biological Macromolecules | 2018
Yujia Liu; Yanxue Cai; DanYang Ying; Yinlian Fu; Ya-Hong Xiong; Xue-Yi Le
Egg ovalbumin (OVA) as a macromolecular carrier has the potential to improve the solubility and stability of insolubility bioactive molecules, however, their binding behavior and the mechanism is still ambiguous. In this work, the curcumin was selected as the target to study the interaction and binding mechanism between curcumin and OVA by thermodynamic titration technique in combination with molecular dynamic simulation. The results suggested that the binding included two steps: first, curcumin molecule entered into the hydrophobic pocket of OVA by hydrophobic interaction; and second the interaction was enhanced via hydrogen bonds, resulting in static fluorescence quenching and secondary structural change of OVA. This study provided further evidence in support of the proposed mechanism of the polyphenol-protein binding by the Hands-gloves model. Furthermore, when the OVA was as a carrier, the solubility of curcumin has been increased ~370 times to 32.73u202fμg/mL compared to that of free curcumin at pHu202f7.0. The photostability was enhanced significantly indicating that it is an efficient way to improve the stability of curcumin in contributing to its application in nutritional supplements or functional foods.