Network


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

Hotspot


Dive into the research topics where Shuai Tang is active.

Publication


Featured researches published by Shuai Tang.


Cancer Cell | 2016

Feedback Activation of Leukemia Inhibitory Factor Receptor Limits Response to Histone Deacetylase Inhibitors in Breast Cancer

Hanlin Zeng; Jia Qu; Nan Jin; Jun Xu; Chenchu Lin; Yi Chen; Xinying Yang; Xiang He; Shuai Tang; Xiaojing Lan; Xiaotong Yang; Ziqi Chen; Min Huang; Jian Ding; Meiyu Geng

Histone deacetylase (HDAC) inhibitors have demonstrated clinical benefits in subtypes of hematological malignancies. However, the efficacy of HDAC inhibitors in solid tumors remains uncertain. This study takes breast cancer as a model to understand mechanisms accounting for limited response of HDAC inhibitors in solid tumors and to seek combination solutions. We discover that feedback activation of leukemia inhibitory factor receptor (LIFR) signaling in breast cancer limits the response to HDAC inhibition. Mechanistically, HDAC inhibition increases histone acetylation at the LIFR gene promoter, which recruits bromodomain protein BRD4, upregulates LIFR expression, and activates JAK1-STAT3 signaling. Importantly, JAK1 or BRD4 inhibition sensitizes breast cancer to HDAC inhibitors, implicating combination inhibition of HDAC with JAK1 or BRD4 as potential therapies for breast cancer.


Cancer Research | 2015

JX06 Selectively Inhibits Pyruvate Dehydrogenase Kinase PDK1 by a Covalent Cysteine Modification

Wenyi Sun; Zuoquan Xie; Yifu Liu; Dan Zhao; Zhixiang Wu; Dadong Zhang; Hao Lv; Shuai Tang; Nan Jin; Hualiang Jiang; Minjia Tan; Jian Ding; Cheng Luo; Jian Li; Min Huang; Meiyu Geng

Pyruvate dehydrogenase kinase PDK1 is a metabolic enzyme responsible for switching glucose metabolism from mitochondrial oxidation to aerobic glycolysis in cancer cells, a general hallmark of malignancy termed the Warburg effect. Herein we report the identification of JX06 as a selective covalent inhibitor of PDK1 in cells. JX06 forms a disulfide bond with the thiol group of a conserved cysteine residue (C240) based on recognition of a hydrophobic pocket adjacent to the ATP pocket of the PDK1 enzyme. Our investigations of JX06 mechanism suggested that covalent modification at C240 induced conformational changes at Arginine 286 through Van der Waals forces, thereby hindering access of ATP to its binding pocket and in turn impairing PDK1 enzymatic activity. Notably, cells with a higher dependency on glycolysis were more sensitive to PDK1 inhibition, reflecting a metabolic shift that promoted cellular oxidative stress and apoptosis. Our findings offer new mechanistic insights including how to therapeutically target PDK1 by covalently modifying the C240 residue.


Journal of Medicinal Chemistry | 2017

Development of the First Generation of Disulfide-Based Subtype-Selective and Potent Covalent Pyruvate Dehydrogenase Kinase 1 (PDK1) Inhibitors

Yifu Liu; Zuoquan Xie; Dan Zhao; Jin Zhu; Fei Mao; Shuai Tang; Hui Xu; Cheng Luo; Meiyu Geng; Min Huang; Jian Li

Pyruvate dehydrogenase kinases (PDKs) are overexpressed in most cancer cells and are responsible for aberrant glucose metabolism. We previously described bis(4-morpholinyl thiocarbonyl)-disulfide (JX06, 16) as the first covalent inhibitor of PDK1. Here, on the basis of the scaffold of 16, we identify two novel types of disulfide-based PDK1 inhibitors. The most potent analogue, 3a, effectively inhibits PDK1 both at the molecular (kinact/Ki = 4.17 × 103 M-1 s-1) and the cellular level (down to 0.1 μM). In contrast to 16, 3a is a potent and subtype-selective inhibitor of PDK1 with >40-fold selectivity for PDK2-4. 3a also significantly alters glucose metabolic pathways in A549 cells by decreasing ECAR and increasing ROS. Moreover, in the xenograft models, 3a shows significant antitumor activity with no negative effect to the mice weight. Collectively, these data demonstrate that 3a may be an excellent lead compound for the treatment of cancer as a first-generation subtype-selective and covalent PDK1 inhibitor.


Organic and Biomolecular Chemistry | 2016

One-pot N-glycosylation remodeling of IgG with non-natural sialylglycopeptides enables glycosite-specific and dual-payload antibody–drug conjugates

Feng Tang; Yang Yang; Yubo Tang; Shuai Tang; Liyun Yang; Bingyang Sun; Bofeng Jiang; Jinhua Dong; Hong Liu; Min Huang; Meiyu Geng; Wei Huang

Chemoenzymatic transglycosylation catalyzed by endo-S mutants is a powerful tool for in vitro glycoengineering of therapeutic antibodies. In this paper, we report a one-pot chemoenzymatic synthesis of glycoengineered Herceptin using an egg-yolk sialylglycopeptide (SGP) substrate. Combining this one-pot strategy with novel non-natural SGP derivatives carrying azido or alkyne tags, glycosite-specific conjugation was enabled for the development of new antibody-drug conjugates (ADCs). The site-specific ADCs and semi-site-specific dual-drug ADCs were successfully achieved and characterized with SDS-PAGE, intact antibody or ADC mass spectrometry analysis, and PNGase-F digestion analysis. Cancer cell cytotoxicity assay revealed that small-molecule drug release of these ADCs relied on the cleavable Val-Cit linker fragment embedded in the structure. These results represent a new approach for glycosite-specific and dual-drug ADC design and rapid synthesis, and also provide the structural requirement for their biologic activities.


Oncogene | 2017

Phosphoglycerate mutase 1 promotes cancer cell migration independent of its metabolic activity

Dadong Zhang; Nan Jin; Wenyi Sun; X Li; B Liu; Zuoquan Xie; Jia Qu; Jun Xu; Xinying Yang; Yi Su; Shuai Tang; H Han; D Chen; Jian Ding; Minjia Tan; Min Huang; Meiyu Geng

Phosphoglycerate mutase 1 (PGAM1) is a glycolytic enzyme that coordinates glycolysis and biosynthesis to promote cancer growth via its metabolic activity. Here, we report the discovery of a non-metabolic function of PGAM1 in promoting cancer metastasis. A proteomic study identified α-smooth muscle actin (ACTA2) as a PGAM1-associated protein. PGAM1 modulated actin filaments assembly, cell motility and cancer cell migration via directly interacting with ACTA2, which was independent of its metabolic activity. The enzymatically inactive H186R mutant retained its association with ACTA2, whereas 201–210 amino acids deleted PGAM1 mutant lost the interaction with ACTA2 regardless of intact metabolic activity. Importantly, PGAM1 knockdown decreased metastatic potential of breast cancer cells in vivo and PGAM1 and ACTA2 were jointly associated with the prognosis of breast cancer patients. Together, this study provided the first evidence revealing a non-metabolic function of PGAM1 in promoting cell migration, and gained new insights into the role of PGAM1 in cancer progression.


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery and structure activity relationship study of novel indazole amide inhibitors for extracellular signal-regulated kinase1/2 (ERK1/2).

Lei Li; Feifei Liu; Nan Jin; Shuai Tang; Zhuxi Chen; Xiaotong Yang; Jian Ding; Meiyu Geng; Lei Jiang; Min Huang; Jianhua Cao

The discovery and optimization of a series of indazole amide based extracellular signal-regulated kinase inhibitors via structure/knowledge based drug design and kinase screen is reported. The optimized compounds demonstrate potent inhibition of ERK1/2 enzyme activity, growth of BRAF mutant HT29 cells and ERK signaling in HT29 cells.


Frontiers in Pharmacology | 2017

Identification of Epigallocatechin-3- Gallate as an Inhibitor of Phosphoglycerate Mutase 1

Xiaoguang Li; Shuai Tang; Qian-Qian Wang; Elaine L.-H. Leung; Hongyue Jin; Yongzhuo Huang; Jia Liu; Meiyu Geng; Min Huang; Shengtao Yuan; Xiao-Jun Yao; Jian Ding

Targeting metabolic enzymes is believed to provide new therapeutic opportunities for cancer therapy. Phosphoglycerate mutase 1 (PGAM1) is a glycolytic enzyme that importantly coordinates glycolysis, pentose phosphate pathway (PPP) flux and serine biosynthesis in cancer cells and hence gains increasing interest of inhibitor discovery. Only few PGAM1 inhibitors have been reported and the molecular potency remains very limited. In an effort to discover new PGAM1 inhibitors, we carried out a biochemical assay-based screen that was focused on natural products derived small molecule compounds. (-)-Epigallocatechin-3-gallate (EGCG), the major natural catechins of green tea extract, was identified as a PGAM1 inhibitor that was tremendously more potent than known PGAM1 inhibitors. Further studies combining molecular docking and site-specific mutagenesis revealed that EGCG inhibited PGAM1 enzymatic activity in a manner independent of substrate competition. EGCG modulated the intracellular level of 2-phosphoglycerate, impaired glycolysis and PPP and inhibited proliferation of cancer cells. This study suggested EGCG as a chemical scaffold for the discovery of potent PGAM1 inhibitors and gained mechanistic insights to understand the previously appreciated anticancer properties of EGCG.


Bioorganic & Medicinal Chemistry Letters | 2017

Discovery of a class of diheteroaromatic amines as orally bioavailable CDK1/4/6 inhibitors

Yan Fu; Shuai Tang; Yi Su; Xiaojing Lan; Yan Ye; Chuantao Zha; Lei Li; Jianhua Cao; Yi Chen; Lei Jiang; Ying Huang; Jian Ding; Meiyu Geng; Min Huang; Huixin Wan

The discovery of a class of diheteroaromatic amines based on LY2835219 as cyclin-dependent kinase (CDK1/4/6) inhibitors was described. The series was found to have much more improved CDK1 inhibition and potent in vitro anti-proliferative effects against cancer cell lines. The synthesis and structure-activity relationship studies of these compounds were reported. One promising compound was selected to evaluate as a novel lead compound after in vitro and in vivo profiling.


Bioorganic & Medicinal Chemistry Letters | 2018

Discovery and structure-activity-relationship study of novel imidazole cyclopropyl amine analogues for mutant isocitric dehydrogenase 1 (IDH1) inhibitors

Qiangang Zheng; Ziqi Chen; Huixin Wan; Shuai Tang; Yan Ye; Yuan Xu; Lei Jiang; Jian Ding; MeiyuGeng; Min Huang; Ying Huang

The discovery and optimization of imidazole cyclopropyl amime analogues as mutant IDH1 inhibitors via structure-based rational design were reported. The optimal compounds demonstrated both potent inhibition in IDH1R132H enzymatic activity and 2HG production in IDH1 mutant HT1080 cell line, moderate liver microsome stability and PK properties.


Cancer Research | 2015

Abstract 1752: 6-Methoxyethylamino-numonafide (MEAN) inhibits hepatocellular carcinoma as a single agent or in combination with sorafenib

Yanning Liu; Guohua Lou; John Norton; Chen Wang; Irawati Kandela; Shuai Tang; Min Huang; Michael J. Avram; Richard M. Green; Andrew P. Mazar; Daniel H. Appella; Zhi Chen; Sui Huang

Hepatocellular carcinoma (HCC) is the third leading form of cancer worldwide and the incidence is increasing rapidly in the United States, tripling over the past 3 decades. Unfortunately, chemotherapeutic treatment strategies against localized and metastatic HCC are very ineffective, leading to a high mortality from the disease. Sorafenib is the sole FDA approved chemotherapeutic currently used clinically for the disease and it shows limited efficacy and substantial toxicity. We have developed a small molecule, 6-methoxyethylamino-numonafide (MEAN), which is highly effective in two murine xenograph models of human HCC. MEAN is more effective in tumor growth inhibition and less toxic than sorafenib at the same concentration. MEAN, at efficacious doses, does not significantly affect animal body weight and does not significantly induce liver damage as determined by the serum levels of liver enzymes at the experimental endpoint. NCI60 cell assay analyses using the COMPARE algorithm indicate that, at IC50 concentration, MEAN does not significantly correlate with any of the small molecules in the 60 cell assay database (correlation Citation Format: Yanning Liu, Guohua Lou, John Norton, Chen Wang, Irawati Kandela, Shuai Tang, Min Huang, Michael Avram, Richard Green, Andrew Mazar, Daniel Appella, Zhi Chen, Sui Huang. 6-Methoxyethylamino-numonafide (MEAN) inhibits hepatocellular carcinoma as a single agent or in combination with sorafenib. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1752. doi:10.1158/1538-7445.AM2015-1752

Collaboration


Dive into the Shuai Tang's collaboration.

Top Co-Authors

Avatar

Min Huang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Meiyu Geng

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Jian Ding

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Nan Jin

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Xiaojing Lan

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yi Su

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

耿美玉

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yan Ye

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yi Chen

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Zhuxi Chen

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge