HuaLiang Jiang

Find an error

Name: 蒋华良; HuaLiang Jiang
Organization: Shanghai Tech University
Department:
Title: Professor
Co-reporter:Diliang Guo, Jing Li, Henry Lin, Yu Zhou, Ying Chen, Fei Zhao, Haifeng Sun, Dan Zhang, Honglin Li, Brian K. Shoichet, Lei Shan, Weidong Zhang, Xin Xie, Hualiang Jiang, and Hong Liu
Journal of Medicinal Chemistry 2016 Volume 59(Issue 20) pp:9489-9502
Publication Date(Web):October 6, 2016
DOI:10.1021/acs.jmedchem.6b01217
A novel series of tetrahydroprotoberberine derivatives (THPBs) were designed, synthesized, and evaluated as selective α1A-adrenergic receptors (AR) antagonists for the treatment of benign prostatic hyperplasia. On the basis of the pharmacophore model of the marketed drug silodosin, THPBs were modified by introducing an indole segment into their core scaffolds. In calcium assays, 7 out of 32 compounds displayed excellent antagonistic activities against α1A-ARs, with IC50 less than 250 nM. Among them, compound (S)-27 had the most potent biological activity; its IC50 toward α1A-AR was 12.8 ± 2.2 nM, which is 781 and 20 times more selective than that toward α1B- and α1D-AR, respectively. In the functional assay using isolated rat tissues, compound (S)-27 inhibited norepinephrine-induced urethra smooth muscle contraction potently (IC50 = 0.5 ± 0.3 nM), without inhibiting the aortic contraction (IC50 > 1000 nM), displaying a better tissue selectivity than the marketed drug silodosin. Additional results of preliminary safety studies (acute toxicity and hERG inhibition) and pharmacokinetics studies indicated the potential druggability for compound (S)-27 which is a promising lead for the development of selective α1A-AR antagonists for the treatment of BPH.
Co-reporter:Shijie Chen; Linjuan Li; Yantao Chen; Junchi Hu; Jingqiu Liu; Yu-Chih Liu; Rongfeng Liu; Yuanyuan Zhang; Fanwang Meng; Kongkai Zhu; Junyan Lu; Mingyue Zheng; Kaixian Chen; Jin Zhang; Hualiang Jiang; Zhiyi Yao;Cheng Luo
Journal of Chemical Information and Modeling 2016 Volume 56(Issue 3) pp:527-534
Publication Date(Web):February 25, 2016
DOI:10.1021/acs.jcim.5b00738
Histone methyltransferases are involved in many important biological processes, and abnormalities in these enzymes are associated with tumorigenesis and progression. Disruptor of telomeric silencing 1-like (DOT1L), a key hub in histone lysine methyltransferases, has been reported to play an important role in the processes of mixed-lineage leukemia (MLL)-rearranged leukemias and validated to be a potential therapeutic target. In this study, we identified a novel DOT1L inhibitor, DC_L115 (CAS no. 1163729-79-0), by combining structure-based virtual screening with biochemical analyses. This potent inhibitor DC_L115 shows high inhibitory activity toward DOT1L (IC50 = 1.5 μM). Through a process of surface plasmon resonance-based binding assays, DC_L115 was founded to bind to DOT1L with a binding affinity of 0.6 μM in vitro. Moreover, this compound selectively inhibits MLL-rearranged cell proliferation with an IC50 value of 37.1 μM. We further predicted the binding modes of DC_L115 through molecular docking analysis and found that the inhibitor competitively occupies the binding site of S-adenosylmethionine. Overall, this study demonstrates the development of potent DOT1L inhibitors with novel scaffolds.
Co-reporter:Hanqi Liu;Linlin Yang;Qiansen Zhang;Lingxue Mao
European Biophysics Journal 2016 Volume 45( Issue 6) pp:511-521
Publication Date(Web):2016 September
DOI:10.1007/s00249-016-1118-1
Caveolin-1 is the principle membrane protein of caveolae and plays an important role in various cellular processes. The protein contains two helices (H1 and H2) connected by a three-residue break. Although caveolin-1 is assumed to adopt a U-shaped conformation in the transmembrane domain, with both the N-terminus and C-terminus exposed to the cytoplasm, the structure and dynamics of caveolin-1 in membranes are still unclear. Here, we performed six molecular dynamics simulations to characterize the structure and dynamics of caveolin-1 (residues D82–S136; Cav182–136) in a caveolae-mimicking asymmetric lipid bilayer. The simulations reveal that the structure of the caveolin scaffolding domain of caveolin-1 is dynamic, as it could be either fully helical or partly unstructured. Cav182–136 inserts into the inner leaflet of the asymmetric lipid bilayer with a stable U-shaped conformation and orients almost vertical to the bilayer surface. The simulations also provide new insights into the effects of caveolin-1 on the morphology of caveolae and the possible interacting site of cholesterol on caveolin-1.
(3-(2,6-Dimethylphenyl)-5-methylisoxazol-4-yl)methanol
3H-Pyrazol-3-one, 1-acetyl-1,2-dihydro-
1H-Pyrazole, 3-(phenylmethoxy)-
2,6-bis(phenylmethoxy)benzamide
2H-Pyrido[3,2-b]-1,4-oxazin-3(4H)-one,6-[[5-fluoro-2-[(3,4,5-trimethoxyphenyl)amino]-4-pyrimidinyl]amino]-2,2-dimethyl-, monobenzenesulfonate
BENZAMIDE, 2,4-BIS(PHENYLMETHOXY)-
(3-(2-Chlorophenyl)-5-methylisoxazol-4-yl)methanol
4-(Benzyloxy)-3-bromo-5-ethoxybenzaldehyde