Network


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

Hotspot


Dive into the research topics where Suhong Yu is active.

Publication


Featured researches published by Suhong Yu.


Scientific Reports | 2015

Nitric Oxide Inhibits Hetero-adhesion of Cancer Cells to Endothelial Cells: Restraining Circulating Tumor Cells from Initiating Metastatic Cascade

Yusheng Lu; Ting Yu; Haiyan Liang; Jichuang Wang; Jingjing Xie; Jingwei Shao; Yu Gao; Suhong Yu; Shuming Chen; Lie Wang; Lee Jia

Adhesion of circulating tumor cells (CTCs) to vascular endothelial bed becomes a crucial starting point in metastatic cascade. We hypothesized that nitric oxide (NO) may prevent cancer metastasis from happening by its direct vasodilation and inhibition of cell adhesion molecules (CAMs). Here we show that S-nitrosocaptopril (CAP-NO, a typical NO donor) produced direct vasorelaxation that can be antagonized by typical NO scavenger hemoglobin and guanylate cyclase inhibitor. Cytokines significantly stimulated production of typical CAMs by the highly-purified human umbilical vein endothelial cells (HUVECs). CAP-NO inhibited expression of the stimulated CAMs (particularly VCAM-1) and the resultant hetero-adhesion of human colorectal cancer cells HT-29 to the HUVECs in a concentration-dependent manner. The same concentration of CAP-NO, however, did not significantly affect cell viability, cell cycle and mitochondrial membrane potential of HT-29, thus excluding the possibility that inhibition of the hetero-adhesion was caused by cytotoxicity by CAP-NO on HT-29. Hemoglobin reversed the inhibition of CAP-NO on both the hetero-adhesion between HT-29 and HUVECs and VCAM-1 expression. These data demonstrate that CAP-NO, by directly releasing NO, produces vasorelaxation and interferes with hetero-adhesion of cancer cells to vascular endothelium via down-regulating expression of CAMs. The study highlights the importance of NO in cancer metastatic prevention.


Cancer | 2015

Isolation and characterization of living circulating tumor cells in patients by immunomagnetic negative enrichment coupled with flow cytometry

Yusheng Lu; Haiyan Liang; Ting Yu; Jingjing Xie; Shuming Chen; Haiyan Dong; Patrick J. Sinko; Shu Lian; Jianguo Xu; Jichuang Wang; Suhong Yu; Jingwei Shao; Bo Yuan; Lie Wang; Lee Jia

This study was aimed at establishing a sensitive and specific isolation, characterization, and enumeration method for living circulating tumor cells (CTCs) in patients with colorectal carcinoma.


Medicinal Research Reviews | 2014

The Unique Pharmacological Characteristics of Mifepristone (RU486): From Terminating Pregnancy to Preventing Cancer Metastasis

Jianzhong Chen; Jichuang Wang; Jingwei Shao; Yu Gao; Jianguo Xu; Suhong Yu; Zhenhua Liu; Lee Jia

Mifepristone (RU486) is a born‐for‐woman molecule discovered three decades ago. Unlike those antihypertensive and antipsychotic pharmaceutical blockbusters, this abortifacient offers relatively low profit potential. Current understanding of mechanism of action of mifepristone and its on‐going clinical trials are changing our views on the drug beyond its abortifacient scope. Here we briefly review its metabolism and pharmacokinetic properties including its unique enterohepatic circulation, its mechanisms of actions involving antiprogesterone and antiglucocorticoid, growth inhibition of various cancer cell lines, suppression of invasive and metastatic cancer potential, downregulation of Cdk2, Bcl‐2, and NF‐kappa B, interference of heterotypic cell adhesion to basement membrane, and cell migration. We comprehensively analyze recent results from preclinical and clinical studies using mifepristone as an anticancer drug for breast, meningioma, and gliomas tumors in the central nervous system, prostate cancer, ovarian and endometrial cancer, and gastric adenocarcinoma. Although mifepristone has more benefits for global public health than we originally thought, its effect as a postmetastatic chemotherapeutic agent is limited. Nonetheless, owing to its unique safe, metabolism and other pharmacological properties, metapristone (the primary metabolite of mifepristone) may have potential for cancer metastatic chemoprevention.


Aaps Journal | 2014

Synthesis, Spectral Characterization, and In Vitro Cellular Activities of Metapristone, a Potential Cancer Metastatic Chemopreventive Agent Derived from Mifepristone (RU486)

Jichuang Wang; Jianzhong Chen; Liyuan Wan; Jingwei Shao; Yusheng Lu; Yewei Zhu; Minrui Ou; Suhong Yu; Haijun Chen; Lee Jia

Mifepristone (RU486) is marketed and used widely by women as an abortifacient, and experimentally for psychotic depression and anticancer treatments. After administration, metapristone is found to be the most predominant metabolite of mifepristone. We hypothesized that adhesion of circulating tumor cells (CTCs) to vascular endothelial bed is a crucial starting point in metastatic cascade, and that metapristone can serve as a cancer metastatic chemopreventive agent that can interrupt adhesion and invasion of CTCs to the intima of microvasculature. In the present study, we modified the synthesis procedure to produce grams of metapristone, fully characterized its spectral properties and in vitro cellular activities, including its cytostatic effects, cell cycle arrest, mitochondrial membrane potential, and apoptosis on human colorectal cancer HT-29 cells. Metapristone concentration dependently interrupted adhesion of HT-29 cells to endothelial cells. Metapristone may potentially be a useful agent to interrupt metastatic initiation.


Biochemical Pharmacology | 2015

UP12, a novel ursolic acid derivative with potential for targeting multiple signaling pathways in hepatocellular carcinoma

Haiyan Dong; Xiang Yang; Jingjing Xie; Liping Xiang; Yuanfang Li; Minrui Ou; Ting Chi; Zhenhua Liu; Suhong Yu; Yu Gao; Jianzhong Chen; Jingwei Shao; Lee Jia

Targeting cancer cell glucose metabolism is a promising strategy for cancer therapy. In past approaches to cancer drug discovery, ursolic acid (UA) has been chemically modified to improve its antitumor activities and bioavailability. Here, a novel ursolic acid (UA) derivative UP12 was developed via computer-aided drug design to explore potent anti-cancer agents and to examine possible mechanisms. The structural docking analyses suggested that UP12 could bind to the active sites of glucokinase (GK), glucose transporter 1 (GLUT1) and ATPase, which are the main enzymes involved in cancer glucose metabolism. We further investigated the synergistic effect between UP12 and glycolysis inhibitor 2-deoxy-d-glucose (2-DG) in inhibiting glucose metabolism of cancer cells. The pharmacological results showed that the combination enhanced depletion of intracellular ATP and decrease in lactate production, and pushed more cancer cells arrested in the S and G2/M cycle phases. The combination selectively down-regulated the expression of Bcl-2 and HKII proteins, up-regulated the expression of Bax and p53, and collectively resulted in enhanced apoptosis related to caspase-3, -8, and -9 activities, in addition to inhibition on the cell mitochondrial membrane potential. The animal studies further demonstrated that the combination exhibited significant antitumor activity without obvious toxicity. In summary, UP12 can interfere cancer cell metabolism pathway and further enhance the therapeutic effects of 2-DG likely through synergistic suppression of cancer cell glucose metabolism, making UP12 a likely new candidate for anti-cancer drug development.


Drug Discovery Today | 2014

Drug enterohepatic circulation and disposition: constituents of systems pharmacokinetics

Yu Gao; Jingwei Shao; Zhou Jiang; Jianzhong Chen; Songen Gu; Suhong Yu; Ke Zheng; Lee Jia

Drug disposition information constitutes a part of systems pharmacokinetics, and becomes imperative when a drug shows significant effects at its disproportionally low blood concentration. The situation could result from outweighing the parent drug in tissues over in blood and/or from its active metabolites. Fractions of certain drugs absorbed from the intestine to the systemic circulation via the portal vein can return to the intestine via the bile duct and the sphincter of Oddi - a complementary nonrenal elimination route termed the enterohepatic circulation (EHC). Here, we critically evaluate the existing methods, techniques and animal models used for determining drug distribution, elimination and EHC, and collectively portray characteristics of 43 drugs that undergo EHC. EHC could represent an unexplored way to excrete unwanted substrates out of the body. The interdisciplinary analysis galvanizes our efforts to overcome technical gaps in drug discovery and development.


Scientific Reports | 2015

Systems pharmacology of mifepristone (RU486) reveals its 47 hub targets and network: Comprehensive analysis and pharmacological focus on FAK-Src-Paxillin complex

Suhong Yu; Xingtian Yang; Yewei Zhu; Fangwei Xie; Yusheng Lu; Ting Yu; Cuicui Yan; Jingwei Shao; Yu Gao; Fan Mo; Guoneng Cai; Patrick J. Sinko; Lee Jia

Mifepristone (RU486), a synthetic steroid compound used as an abortifacient drug, has received considerable attention to its anticancer activity recently. To explore the possibility of using mifepristone as a cancer metastasis chemopreventive, we performed a systems pharmacology analysis of mifepristone-related molecules in the present study. Data were collected by using Natural Language Processing (NLP) and 513 mifepristone-related genes were dug out and classified functionally using a gene ontology (GO) hierarchy, followed by KEGG pathway enrichment analysis. Potential signal pathways and targets involved in cancer were obtained by integrative network analysis. Total thirty-three proteins were involved in focal adhesion-the key signaling pathway associated with cancer metastasis. Molecular and cellular assays further demonstrated that mifepristone had the ability to prevent breast cancer cells from migration and interfere with their adhesion to endothelial cells. Moreover, mifepristone inhibited the expression of focal adhesion kinase (FAK), paxillin, and the formation of FAK/Src/Paxillin complex, which are correlated with cell adhesion and migration. This study set a good example to identify chemotherapeutic potential seamlessly from systems pharmacology to cellular pharmacology, and the revealed hub genes may be the promising targets for cancer metastasis chemoprevention.


Journal of Pharmaceutical and Biomedical Analysis | 2015

Bioactivity-guided fast screen and identification of cancer metastasis chemopreventive components from raw extracts of Murraya exotica.

Zhou Jiang; Jina Yang; Yaqiong Pang; Xintian Yang; Suhong Yu; Lee Jia

Murraya exotica is a traditional Chinese medicine (TCM) widely grown in southeast China. We herein proposed a fast strategy for separation and identification of active components of cancer metastatic chemopreventives from the root, leaf, twig and stem bark extracts that were obtained by reflux in 80% acidic ethanol and then liquid-liquid extraction. High performance liquid chromatography (HPLC) analysis showed that the extract mixtures from leaf, bark and twig were similar, while the root extract contained a characteristic component (CM1). Bioactivity assays revealed that the root extract contained some active components that significantly inhibited cancer cell viability and migration. Ultra performance liquid chromatography coupled with diode array detection and electrospray ionization mass spectrometry (UPLC-DAD-ESI-MS) analysis indicated the existence of coumarins in the root and leaf extracts. Semi-preparative chromatographic separation and physicochemical characterization indicated that CM1 was a novel coumarin derivative that warrants further chemopreventive studies on cancer metastasis. The present phytochemical and phytopharmacological studies exemplify a fast strategy for screening and identifying active component(s) from raw extracts of phytomedicines.


Journal of Controlled Release | 2015

Ex vivo and in vivo capture and deactivation of circulating tumor cells by dual-antibody-coated nanomaterials

Jingjing Xie; Yu Gao; Rongli Zhao; Patrick J. Sinko; Songen Gu; Jichuang Wang; Yuanfang Li; Yusheng Lu; Suhong Yu; Lie Wang; Shuming Chen; Jingwei Shao; Lee Jia

Circulating tumor cells (CTCs) have been detected by us and others in cancer patient blood. However, little is known about how to specifically capture and deactivate CTCs in vivo, which may lead to successful metastasis prevention in asymptomatic cancer survivors after surgery. We hypothesize that the dual antibody conjugates may have the advantage of capturing CTCs specifically over their single antibody counterparts. Here we show that the surface-functionalized dendrimers can be sequentially coated with two antibodies directed to surface biomarkers (EpCAM and Slex) of human colorectal CTCs. The dual antibody-coated dendrimers exhibit a significantly enhanced specificity in capturing CTCs in the presence of interfering blood cells, and in both eight-patient bloods and nude mice administered with the labeled CTCs in comparison to their single antibody-coated counterparts. The dual antibody-coated conjugates down-regulate the captured CTCs. This study provides the first conceptual evidence that two antibodies can be biocompatibly conjugated to a nanomaterial to capture and down-regulate CTCs in vivo with the enhanced specificity.


Scientific Reports | 2016

Pharmacoproteomic analysis reveals that metapristone (RU486 metabolite) intervenes E-cadherin and vimentin to realize cancer metastasis chemoprevention.

Suhong Yu; Cuicui Yan; Xingtian Yang; Sudang He; Jian Liu; Chongtao Qin; Chuanzhong Huang; Yusheng Lu; Zhongping Tian; Lee Jia

Metapristone is the most predominant biological active metabolite of mifepristone, and being developed as a novel cancer metastasis chemopreventive agent by us. Despite its prominent metastasis chemopreventive effect, the underlying mechanism remains elusive. Our study, for the first time, demonstrated that metapristone had the ability to prevent breast cancer cells from migration, invasion, and interfere with their adhesion to endothelial cells. To explore the underlying mechanism of metapristone, we employed the iTRAQ technique to assess the effect of metapristone on MDA-MB-231 cells. In total, 5,145 proteins were identified, of which, 311 proteins showed significant differences in metapristone-treated cells compared to the control group (P-value < 0.05). Bioinformatic analysis showed many differentially expressed proteins (DEPs) functionally associated with post-translational modification, chaperones, translation, transcription, replication, signal transduction, etc. Importantly, many of the DEPs, such as E-cadherin, vimentin, TGF-β receptor I/II, smad2/3, β-catenin, caveolin, and dystroglycan were associated with TGF-β and Wnt signaling pathways, which were also linked to epithelial-to-mesenchymal transition (EMT) process. Further validation of the epithelial marker “E-caderin” and mesenchymal marker “vimetin” were carried out using immunoblot and immunofluorescence. These results have revealed a novel mechanism that metapristone-mediated metastasis chemoprevention is through intervening the EMT-related signaling pathways.

Collaboration


Dive into the Suhong Yu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianzhong Chen

Fujian University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge