Network


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

Hotspot


Dive into the research topics where Ke-Bin Huang is active.

Publication


Featured researches published by Ke-Bin Huang.


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 | 2013

The antitumor activity of zinc(II) and copper(II) complexes with 5,7-dihalo-substituted-8-quinolinoline.

Yan-Cheng Liu; Jian-Hua Wei; Zhen-Feng Chen; Mei Liu; Yun-Qiong Gu; Ke-Bin Huang; Zhu-Quan Li; Hong Liang

[Zn₂(ClQ)₄(CH₃OH)₂] (1), [Zn(BrQ)₂(H₂O)₂] (2), [Zn₂(ClIQ)₄] (3) and [Cu(BrQ)₂] (4) (H-ClQ = 5,7-dichloro-8-hydroxylquinoline, H-BrQ = 5,7-dibromo-8-hydroxylquinoline, and H-ClIQ = 5-chloro-7-iodo-8-hydroxylquinoline) were synthesized. Compounds 1-4 showed high anti-proliferative cytotoxicities against BEL-7404, SK-OV-3, NCI-H460 tumor cells, and HL-7702 normal cells in vitro, with IC₅₀ values in the 1.4 nM to 32.13 μM range. Compounds 2-4 exhibited significantly enhanced cytotoxicity against BEL-7404 cell line, comparing with free 5,7-dihalo-8-quinolinol. Western blotting analysis showed that 2, 3 depleted mutant p53 protein in MDA-MB-231, and compound 2 decreased the ratio of Bcl-2/Bax in NCI-H460 significantly. The binding abilities of 1-4 to DNA were stronger than that of free quinolinol ligand. Intercalation is the probable binding mode for the complexes and free quinolinol ligands with DNA.


European Journal of Medicinal Chemistry | 2015

Studies on antitumor mechanism of two planar platinum(II) complexes with 8-hydroxyquinoline: synthesis, characterization, cytotoxicity, cell cycle and apoptosis.

Qi-Pin Qin; Zhen-Feng Chen; Jiao-Lan Qin; Xiao-Ju He; Yu-Lan Li; Yan-Cheng Liu; Ke-Bin Huang; Hong Liang

[Pt(Q)2] (1) and [Pt(MQ)2] (2) exhibited enhanced cytotoxicity against BEL-7404, Hep-G2, NCI-H460, T-24, A549 tumor cells but low cytotoxicity on normal HL-7702 cells. 1 and 2 could cause the cell cycle arrest in G2 and S phase, respectively. While pifithrin-α, a specific p53 inhibitor, induced cell cycle arrest in G1 phase. Although 1, 2 and pifithrin-α caused serious inhibition on p53, 1 and 2 significantly cause the loss of mitochondrial membrane potential and increase of the reactive oxygen species level, cytochrome c, apaf-1 and caspase-3/9 ratio in BEL-7404 cells. 1 and 2 may trigger the cell apoptosis through a mitochondrial dysfunction pathway whereas pifithrin-α does not. The interactions of 1 and 2 with DNA are most probably via an intercalation.


European Journal of Medicinal Chemistry | 2013

High antitumor activity of 5,7-dihalo-8-quinolinolato tin(IV) complexes

Zhen-Feng Chen; Yan Peng; Yun-Qiong Gu; Yan-Cheng Liu; Mei Liu; Ke-Bin Huang; Kun Hu; Hong Liang

Three tin(IV) complexes [Sn(ClQ)2Cl2] (1), [Sn(BrQ)2Cl2] (2) and [Sn(ClIQ)2Cl2] (3) were prepared (H-ClQ = 5,7-dichloro-8-hydroxylquinoline, H-BrQ = 5,7-dibromo-8-hydroxylquinoline, H-ClIQ = 5-chloro-7-iodo-8-hydroxylquinoline) and their in vitro cytotoxicities against BEL7404, SKOV-3, NCI-H460, HL-7702 cell lines were evaluated. The complexes showed high anti-proliferative activity toward the tested cell lines with IC50 values ranging from 20 nM to 5.11 μM. Compared with 5,7-dihalo-8-quinolinol, most complexes exhibited significantly enhanced cytotoxicity (except 2 against SKOV-3 and NCI-H460). They also displayed some selective cytotoxicity favoring the tested tumor cells over the normal human liver HL-7702 cells. Compared with their quinolinol ligands, complexes 1-3 bind more strongly with DNA. Intercalation is the most probable binding mode for both the complexes and their quinolinol ligands.


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.


Journal of Inorganic Biochemistry | 2017

Crystal structure, cytotoxicity and action mechanism of Zn(II)/Mn(II) complexes with isoquinoline ligands

Feng-Yang Wang; Qian-Yu Xi; Ke-Bin Huang; Xiao-Ming Tang; Zhen-Feng Chen; Yan-Cheng Liu; Hong Liang

Four μ2-Cl bridged dinuclear metal complexes with isoquinoline ligands, (MPDQ)2Zn2Cl4 (1) (MPDQ=4.5-methylenedioxy-1-pyridinedihydroisoquinoline), (PYP)2Zn2Cl4 (2) (PYP=5-pyridin-2-yl-[1,3]dioxolo[4,5-g]isoquinoline), (MPDQ)2Mn2Cl4 (3),and (PYP)2Mn2Cl4 (4) were synthesized and characterized. All complexes exhibited strong proliferation inhibition activity against various cancer cells. The underlying molecular mechanisms through which they caused the cancer cell death were also elucidated. Induction of apoptosis in MGC-803 cells by complex 2 was evidenced by annexin V+/PI- detection and DiD/DAPI staining assay. Further investigation revealed that complex 2 was able to induce intrinsic pathway-dependent apoptosis in cancer cells by triggering DNA damage which was caused by the overproduction of reactive oxygen species. Based on these studies, we suggest that Zn(II) complexes containing isoquinoline ligands can be developed as candidates for anti-cancer chemotherapeutics.


Journal of Medicinal Chemistry | 2018

Organometallic Gold(III) Complexes Similar to Tetrahydroisoquinoline Induce ER-Stress-Mediated Apoptosis and Pro-Death Autophagy in A549 Cancer Cells

Ke-Bin Huang; Feng-Yang Wang; Xiao-Ming Tang; Hai-Wen Feng; Zhen-Feng Chen; Yan-Cheng Liu; You-Nian Liu; Hong Liang

Agents inducing both apoptosis and autophagic death can be effective chemotherapeutic drugs. In our present work, we synthesized two organometallic gold(III) complexes harboring C^N ligands that structurally resemble tetrahydroisoquinoline (THIQ): Cyc-Au-1 (AuL1Cl2, L1 = 3,4-dimethoxyphenethylamine) and Cyc-Au-2 (AuL2Cl2, L2 = methylenedioxyphenethylamine). In screening their in vitro activity, we found both gold complexes exhibited lower toxicity, lower resistance factors, and better anticancer activity than those of cisplatin. The organometallic gold(III) complexes accumulate in mitochondria and induce elevated ROS and an ER stress response through mitochondrial dysfunction. These effects ultimately result in simultaneous apoptosis and autophagy. Importantly, compared to cisplatin, Cyc-Au-2 exhibits lower toxicity and better anticancer activity in a murine tumor model. To the best of our knowledge, Cyc-Au-2 is the first organometallic Au(III) compound that induces apoptosis and autophagic death. On the basis of our results, we believe Cyc-Au-2 to be a promising anticancer agent or lead compound for further anticancer drug development.


Free Radical Biology and Medicine | 2018

New Platinum(II) agent induces bimodal death of apoptosis and autophagy against A549 cancer cell

Feng-Yang Wang; Ke-Bin Huang; Hai-Wen Feng; Zhen-Feng Chen; You-Nian Liu; Hong Liang

&NA; Agents with multiple modes of tumor cell death can be effective chemotherapeutic drugs. One example of a bimodal chemotherapeutic approach is an agent that can induce both apoptosis and autophagic death. Thus far, no clinical anticancer drug has been shown to simultaneously induce both these pathways. Mono‐functional platinum complexes are potent anticancer drug candidates which act through mechanisms distinct from cisplatin. Here, we describe the synthesis and characterize of two mono‐functional platinum complexes containing 8‐substituted quinoline derivatives as ligands. In comparison to cisplatin, n‐Mon‐Pt‐1 exhibited a greater in vitro cytotoxicity, was more effective in resistant cells and elicited a better anticancer effect. Mechanistic experiments indicate that n‐Mon‐Pt‐1 mainly accumulates in mitochondria, and stimulates significant TrxR inhibition, ROS release and an ER stress response, ultimately resulting in a simultaneous induction of apoptosis and autophagy. Importantly, compared to cisplatin, n‐Mon‐Pt‐1 exhibits lower acute toxicity and better anticancer activity in a murine tumor model. Graphical abstract Figure. No caption available. HighlightsTwo novel mono‐functional platinum complexes were synthesized.The complexes exhibited mitochondria‐targeting.The antitumor activity was resulted from the apoptosis and autophagy simultaneously.


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.


Acta Crystallographica Section E-structure Reports Online | 2010

(Sulfasalazinato-κO)bis-(triphenyl-phosphine-κP)copper(I).

Ke-Bin Huang; Yan-Shu Tan; Xiao-Yan Song; Yan-Cheng Liu; Zhen-Feng Chen

The title mixed-ligand copper(I) complex, [Cu(C18H13N4O5S)(C18H15P)2], was synthesized via solvothermal reaction of [Cu(PPh3)2(MeCN)2]ClO4 and sulfasalazine [systematic name: 2-hydroxy-5-(2-{4-[(2-pyridylamino)sulfonyl]phenyl}diazenyl)benzoic acid]. The mononuclear complex displays a trigonal coordination geometry for the Cu(I) atom, which is surrounded by two P-atom donors from two different PPh3 ligands and one O-atom donor from the monodentate carboxylate group of the sulfasalazinate ligand. The latter ligand is found in a zwitterionic form, with a deprotonated amine N atom and a protonated pyridine N atom. Such a feature was previously described for free sulfasalazine. The crystal structure is stabilized by C—H⋯O, C—H⋯N, N—H⋯N and O—H⋯O hydrogen bonds.

Collaboration


Dive into the Ke-Bin Huang's collaboration.

Top Co-Authors

Avatar

Zhen-Feng Chen

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Hong Liang

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

Feng-Yang Wang

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

Xiao-Ming Tang

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

Qian-Yu Xi

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

Ting Meng

Guangxi Normal University

View shared research outputs
Top Co-Authors

Avatar

You-Nian Liu

Central South University

View shared research outputs
Researchain Logo
Decentralizing Knowledge