Minmin Zhang
Chinese Academy of Sciences
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Minmin Zhang.
Acta Pharmaceutica Sinica B | 2016
Danqi Chen; Aijun Shen; Guanghua Fang; Hongchun Liu; Minmin Zhang; Shuai Tang; Bing Xiong; Lanping Ma; Meiyu Geng; Jingkang Shen
Histone acetylation is a critical process in the regulation of chromatin structure and gene expression. Histone deacetylases (HDACs) remove the acetyl group, leading to chromatin condensation and transcriptional repression. HDAC inhibitors are considered a new class of anticancer agents and have been shown to alter gene transcription and exert antitumor effects. This paper describes our work on the structural determination and structure-activity relationship (SAR) optimization of tetrahydroisoquinoline compounds as HDAC inhibitors. These compounds were tested for their ability to inhibit HDAC 1, 3, 6 and for their ability to inhibit the proliferation of a panel of cancer cell lines. Among these, compound 82 showed the greatest inhibitory activity toward HDAC 1, 3, 6 and strongly inhibited growth of the cancer cell lines, with results clearly superior to those of the reference compound, vorinostat (SAHA). Compound 82 increased the acetylation of histones H3, H4 and tubulin in a concentration-dependent manner, suggesting that it is a broad inhibitor of HDACs.
ACS Medicinal Chemistry Letters | 2017
Dong Lu; Juan Yan; Lang Wang; Hongchun Liu; Limin Zeng; Minmin Zhang; Wenwen Duan; Yinchun Ji; Jingchen Cao; Meiyu Geng; Aijun Shen; Youhong Hu
Simultaneous blockade of more than one pathway is considered to be a promising approach to overcome the low efficacy and acquired resistance of cancer therapies. Thus, a novel series of c-Met/HDAC bifunctional inhibitors was designed and synthesized by merging pharmacophores of c-Met and HDAC inhibitors. The most potent compound, 2m, inhibited c-Met kinase and HDAC1, with IC50 values of 0.71 and 38 nM, respectively, and showed efficient antiproliferative activities against both EBC-1 and HCT-116 cells with greater potency than the reference drug Chidamide. Western blot analysis revealed that compound 2m inhibited phosphorylation of c-Met and c-Met downstream signaling proteins and increased expression of Ac-H3 and p21 in EBC-1 cells in a dose-dependent manner. Our study presents novel compounds for the further exploration of dual c-Met/HDAC pathway inhibition achieved with a single molecule.
Molecular Pharmaceutics | 2016
Liandi Chi; Delin Wu; Zhuo Li; Minmin Zhang; Hongchun Liu; Caifen Wang; Shuangying Gui; Meiyu Geng; Haiyan Li; Jiwen Zhang
In answering to the challenge of enzymatic unstability of Biopharmaceutics Classification System (BCS) class II drugs, an effective remote loading strategy was developed to successfully incorporate the drug-cyclodextrin (CD) complex into niosomes to modify the release and stability of a drug candidate, pseudolaric acid B (PAB). Judged by binding constants, and combined solubilization effects of pH and CD complexation on PAB at different pH, the complex internalization driven by a transmembrane pH gradient (from 2.0 to 7.4) and the dynamic shifting of PAB-CD complexation equilibrium at this gradient were introduced. The transfer of PAB-CD complex into the internal aqueous phase of niosomes at 60 °C was primarily verified by synchrotron radiation Fourier transform infrared spectroscopy. The remote loading samples behaved as retarded release at pH 5.8, 6.8, and 7.4, for which the stability of PAB in rat plasma was significantly enhanced (about 8.1-fold), in comparison with niosomes prepared by the passive and lipid bilayer loading of PAB. The drug-carrier interaction based release modeling was further fitted, and the convection rate constant (ks) and free energy difference between free and bound states (ΔG) indicated the strongest PAB-carrier interactions in remote loading niosomes. The remote loading strategy also reduced the CD-cholesterol interaction and provided better physical stability of the system. In conclusion, the remote loading of drug-CD complex into niosomes provides advantages to modify the release and enhance the stability of unstable BCS class II drug.
Chemical Research in Chinese Universities | 2006
Jing-hui Yan; Minmin Zhang; Hongzhou Lian; J. Liu; Zhi Li; Jie Cao; Congyun Shi
The phase diagram of a cetyltrimethyl ammonium bromide( CTAB)/n-butanol/n-octane/KNO3-Mg( NO3)(2) system was drawn. Nanoparticles of Eu2+-doped KMgF3 were prepared from the quaternary microemulsions of cetyltrimethyl ammonium bromide(CTAB), n-butanol, n-octane and water. The X-ray diffraction(XRD) patterns were indexed to a pure KMgF3 cubic phase. The environmental scanning electron microscopic (ESEM) images show the presence of spherical Eu2+-doped KMgF3 nanoparticles with a diameter of ca. 20 nm. The emission of KMgF3: Eu2+ nanoparticles peaks at 360 mn. The excitation band was observed at 250 nm with a blue shift of ca. 70 nm compared with that of KMgF3: Eu2+ single crystal. The preparation method of nano-KMgF3: Eu2+/PMMA composite films was inquired into.
Acta Pharmacologica Sinica | 2017
Jingya Sun; Jidong Wang; Xin Wang; Hongchun Liu; Minmin Zhang; Yu-Chih Liu; Chen-hua Zhang; Yi Su; Yanyan Shen; Yue-Wei Guo; Aijun Shen; Meiyu Geng
Histone deacetylases (HDACs), especially HDAC1, 2, 3 and 4, are abundantly expressed and over-activated in prostate cancer that is correlated with the poor prognosis. Thus, inhibition of HDAC activity has emerged as a potential alternative option for prostate cancer therapy. Chromopeptide A is a depsipeptide isolated from the marine sediment-derived bacterium Chromobacterium sp. HS-13-94; it has a chemical structure highly similar to FK228, a class I HDAC inhibitor that is approved by FDA for treating T-cell lymphoma. In this study, we determined whether chromopeptide A, like FK228, acted as a class I HDAC inhibitor, and whether chromopeptide A could inhibit the growth and migration of human prostate cancer in vitro and in vivo. HDAC enzyme selectivity and kinetic analysis revealed that chromopeptide A selectively inhibited the enzymatic activities of HDAC1, 2, 3 and 8 in a substrate non-competitive manner with comparable IC50 values for each HDAC member as FK228 in vitro. Importantly, chromopeptide A dose-dependently suppressed the proliferation of human prostate cancer cell lines PC3, DU145 and LNCaP with IC50 values of 2.43±0.02, 2.08±0.16, and 1.75±0.06 nmol/L, respectively, accompanied by dose-dependent inhibition of HDAC enzymatic activity in PC3 and DU145 cells. Chromopeptide A (0.2–50 nmol/L) caused G2/M phase arrest and induced apoptosis in the prostate cancer cell lines. Moreover, chromopeptide A dose-dependently inhibited the migration of PC3 cells. In mice bearing PC3 prostate cancer xenografts, intravenous injection of chromopeptide A (1.6, 3.2 mg/kg, once a week for 18 d) significantly suppressed the tumor growth, which was associated with increased expression levels of Ac-H3 and p21 in tumor tissues. Our results identify chromopeptide A as a novel class I HDAC inhibitor and provide therapeutic strategies that may be implemented in prostate cancer.
Acta Pharmacologica Sinica | 2016
Yueqin Wang; Aijun Shen; Jingya Sun; Xin Wang; Hongchun Liu; Minmin Zhang; Danqi Chen; Bing Xiong; Jingkang Shen; Meiyu Geng; Min Zheng; Jian Ding
Aim:Inhibition of heat shock protein (Hsp90) has been proven to be effective in overriding primary and acquired resistance of kinase inhibitors. In this study, we investigated the role of FS-108, a newly developed Hsp90 inhibitor, to overcome gefitinib resistance in EGFR mutant non-small cell lung cancer cells.Methods:Cell proliferation was assessed using the SRB assay. Cell cycle distribution and apoptosis were analyzed by flow cytometry. Protein expression was examined by Western blotting. The in vivo effectiveness of FS-108 was determined in an NCI-H1975 subcutaneous xenograft model.Results:FS-108 triggered obvious growth inhibition in gefitinib-resistant HCC827/GR6, NCI-H1650 and NCI-H1975 cells through inducing G2/M phase arrest and apoptosis. FS-108 treatment resulted in a remarkable degradation of key client proteins involved in gefitinib resistance and further abrogated their downstream signaling pathways. Interestingly, FS-108 alone exerted an identical or superior effect on circumventing gefitinib resistance compared to combined kinase inhibition. Finally, the ability of FS-108 to overcome gefitinib resistance in vivo was validated in an NCI-H1975 xenograft model.Conclusion:FS-108 is a powerful agent that impacts the survival of gefitinib-resistant cells in vitro and in vivo through targeting Hsp90.
Theranostics | 2018
Jingya Sun; Xin Wang; Boyun Tang; Hongchun Liu; Minmin Zhang; Yueqin Wang; Fangfang Ping; Jian Ding; Aijun Shen; Meiyu Geng
Over the past decade, therapies targeting the VEGF/VEGFR and mTOR pathways have served as the standard of care for the clinical management of renal cell carcinoma (RCC) patients. Albeit promising, these targeted drugs have attained only modest clinical benefits with limited prolonged progression-free survival. Therefore, alternative reasonable and applicable therapeutic approaches should be introduced to improve the clinical outcome of RCC patients. Methods: FDA approved kinase inhibitors were screened to evaluate their abilities to suppress the proliferation of RCC cells. Then, the downstream effector, therapeutic target and signaling pathway of the selected drug were identified by gene expression array, RNAi, kinase profile and rescue verification. Finally, the in vivo effectiveness of the drug was assessed in cell line-based xenograft models and patient-derived xenograft models. Results: In this study, we discovered that dasatinib is a potent agent that can impair RCC cell viability in vitro and decrease tumor growth in vivo. Mechanistically, we improved the understanding of the precise mechanistic role of YAP as a pivotal effector of dasatinib-induced anti-proliferation through Src-JNK-LIMD1-LATS signaling cascade in RCC cells. Meanwhile, our results indicated that the alteration of p-YAP is closely correlated to the growth inhibition caused by dasatinib in sensitive RCC models. Conclusion: Our findings provide evidence that dasatinib may serve as a powerful drug candidate to treat subgroups of RCC patients with hyper-activated Src-YAP signaling axis, and the alteration of p-YAP could serve as a functional response biomarker of dasatinib in RCC.
Hepatology | 2018
Xin Wang; Minmin Zhang; Fangfang Ping; Hongchun Liu; Jingya Sun; Yueqin Wang; Aijun Shen; Jian Ding; Meiyu Geng
Though kinase inhibitors have been heavily investigated in the clinic to combat advanced hepatocellular carcinoma (HCC), clinical outcomes have been disappointing overall, which may be due to the absence of kinase‐addicted subsets in HCC patients. Recently, strategies that simultaneously inhibit multiple kinases are increasingly appreciated in HCC treatment, yet they are challenged by the dynamic nature of the kinase networks. This study aims to identify clustered kinases that may cooperate to drive the malignant growth of HCC. We show that anaplastic lymphoma kinase, fibroblast growth factor receptor 2, and ephrin type‐A receptor 5 are the essential kinases that assemble into a functional cluster to sustain the viability of HCC cells through downstream protein kinase B–dependent, extracellular signal–regulated kinase–dependent, and p38‐dependent signaling pathways. Their coactivation is associated with poor prognosis for overall survival in about 13% of HCC patients. Moreover, their activities are tightly regulated by heat shock protein 90 (Hsp90). Thereby Combined kinase inhibition or targeting of heat shock protein 90 led to significant therapeutic responses both in vitro and in vivo. Conclusion: Our findings established a paradigm that highlights the cooperation of anaplastic lymphoma kinase, fibroblast growth factor receptor 2, and ephrin type‐A receptor 5 kinases in governing the growth advantage of HCC cells, which might offer a conceptual “combined therapeutic target” for diagnosis and subsequent intervention in a subgroup of HCC patients.Though kinase inhibitors are heavily investigated in clinic to combat advanced hepatocellular carcinoma (HCC), clinical outcomes are overall disappointing, which may result from the absence of kinase-addicted subsets in HCC. Recently, a combination strategy that simultaneously inhibits multiple kinases are increasingly appreciated in HCC treatment, yet it is challenged by the dynamic nature of kinase network. This study aims to identify kinase clusters that may cooperate to drive the malignant growth of HCC. We reveal that ALK, FGFR2 and EphA5 are the essential kinases that assembled into a functional cluster to sustain the viability of HCC cells through downstream AKT, ERK and p38 dependent signaling pathways. Their co-activation is associated with the poor prognosis for overall survival in about 13% of HCC patients. Therapeutically, combined inhibition of kinases inhibitors or targeting heat shock protein 90 (Hsp90) led to significant therapeutic response in vitro and in vivo. Conclusion: Our findings revealed a paradigm that highlights the cooperation of ALK, FGFR2 and EphA5 kinases in governing growth advantage of HCC cells, which might offer a novel conceptual “combined therapeutic target” for diagnosis and subsequent intervention in subgroup of HCC patients. Page 4 of 119 Hepatology Hepatology This article is protected by copyright. All rights reserved.
European Journal of Medicinal Chemistry | 2017
Chi Zhang; Xin Wang; Hongchun Liu; Minmin Zhang; Meiyu Geng; Liping Sun; Aijun Shen; Ao Zhang
European Journal of Medicinal Chemistry | 2018
Sumei Ren; Yujie Wang; Junfei Wang; Dingding Gao; Minmin Zhang; Ning Ding; Yingxia Li