Network


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

Hotspot


Dive into the research topics where Yanke Yu is active.

Publication


Featured researches published by Yanke Yu.


Clinical Cancer Research | 2010

Sulforaphane, a Dietary Component of Broccoli/Broccoli Sprouts, Inhibits Breast Cancer Stem Cells

Yanyan Li; Tao Zhang; Hasan Korkaya; Suling Liu; Hsiu Fang Lee; Bryan Newman; Yanke Yu; Shawn G. Clouthier; Steven J. Schwartz; Max S. Wicha; Duxin Sun

Purpose: The existence of cancer stem cells (CSCs) in breast cancer has profound implications for cancer prevention. In this study, we evaluated sulforaphane, a natural compound derived from broccoli/broccoli sprouts, for its efficacy to inhibit breast CSCs and its potential mechanism. Experimental Design: Aldefluor assay and mammosphere formation assay were used to evaluate the effect of sulforaphane on breast CSCs in vitro. A nonobese diabetic/severe combined immunodeficient xenograft model was used to determine whether sulforaphane could target breast CSCs in vivo, as assessed by Aldefluor assay, and tumor growth upon cell reimplantation in secondary mice. The potential mechanism was investigated using Western blotting analysis and β-catenin reporter assay. Results: Sulforaphane (1-5 μmol/L) decreased aldehyde dehydrogenase–positive cell population by 65% to 80% in human breast cancer cells (P < 0.01) and reduced the size and number of primary mammospheres by 8- to 125-fold and 45% to 75% (P < 0.01), respectively. Daily injection with 50 mg/kg sulforaphane for 2 weeks reduced aldehyde dehydrogenase–positive cells by >50% in nonobese diabetic/severe combined immunodeficient xenograft tumors (P = 0.003). Sulforaphane eliminated breast CSCs in vivo, thereby abrogating tumor growth after the reimplantation of primary tumor cells into the secondary mice (P < 0.01). Western blotting analysis and β-catenin reporter assay showed that sulforaphane downregulated the Wnt/β-catenin self-renewal pathway. Conclusions: Sulforaphane inhibits breast CSCs and downregulates the Wnt/β-catenin self-renewal pathway. These findings support the use of sulforaphane for the chemoprevention of breast cancer stem cells and warrant further clinical evaluation. Clin Cancer Res; 16(9); 2580–90. ©2010 AACR.


Biochemical Pharmacology | 2010

Withaferin A targets heat shock protein 90 in pancreatic cancer cells.

Yanke Yu; Adel Hamza; Tao Zhang; Mancang Gu; Peng Zou; Bryan Newman; Yanyan Li; A. A. Leslie Gunatilaka; Chang-Guo Zhan; Duxin Sun

The purpose of this study is to investigate the efficacy and the mechanism of Hsp90 inhibition of Withaferin A (WA), a steroidal lactone occurring in Withania somnifera, in pancreatic cancer in vitro and in vivo. Withaferin A exhibited potent antiproliferative activity against pancreatic cancer cells in vitro (with IC(50)s of 1.24, 2.93 and 2.78 microM) in pancreatic cancer cell lines Panc-1, MiaPaCa2 and BxPc3, respectively. Annexin V staining showed that WA induced significant apoptosis in Panc-1 cells in a dose-dependent manner. Western blotting demonstrated that WA inhibited Hsp90 chaperone activity to induce degradation of Hsp90 client proteins (Akt, Cdk4 and glucocorticoid receptor), which was reversed by the proteasomal inhibitor, MG132. WA-biotin pull down assay of Hsp90 using Panc-1 cancer cell lysates and purified Hsp90 showed that WA-biotin binds to C-terminus of Hsp90 which was competitively blocked by unlabeled WA. Co-immunoprecipitation exhibited that WA (10 microM) disrupted Hsp90-Cdc37 complexes from 1 to 24h post-treatment, while it neither blocked ATP binding to Hsp90, nor changed Hsp90-P23 association. WA (3, 6mg/kg) inhibited tumor growth in pancreatic Panc-1 xenografts by 30% and 58%, respectively. These data demonstrate that Withaferin A binds Hsp90, inhibits Hsp90 chaperone activity through an ATP-independent mechanism, results in Hsp90 client protein degradation, and exhibits in vivo anticancer activity against pancreatic cancer.


Journal of Biological Chemistry | 2009

Characterization of celastrol to inhibit hsp90 and cdc37 interaction.

Tao Zhang; Yanyan Li; Yanke Yu; Peng Zou; Yiqun Jiang; Duxin Sun

The molecular chaperone heat shock protein 90 (Hsp90) is required for the stabilization and conformational maturation of various oncogenic proteins in cancer. The loading of protein kinases to Hsp90 is actively mediated by the cochaperone Cdc37. The crucial role of the Hsp90-Cdc37 complex has made it an exciting target for cancer treatment. In this study, we characterize Hsp90 and Cdc37 interaction and drug disruption using a reconstituted protein system. The GST pull-down assay and ELISA assay show that Cdc37 binds to ADP-bound/nucleotide-free Hsp90 but not ATP-bound Hsp90. Celastrol disrupts Hsp90-Cdc37 complex formation, whereas the classical Hsp90 inhibitors (e.g. geldanamycin) have no effect. Celastrol inhibits Hsp90 ATPase activity without blocking ATP binding. Proteolytic fingerprinting indicates celastrol binds to Hsp90 C-terminal domain to protect it from trypsin digestion. These data suggest that celastrol may represent a new class of Hsp90 inhibitor by modifying Hsp90 C terminus to allosterically regulate its chaperone activity and disrupt Hsp90-Cdc37 complex.


Molecular Pharmaceutics | 2010

Superparamagnetic iron oxide “nanotheranostics” for targeted cancer cell imaging and pH-dependent intracellular drug release

Peng Zou; Yanke Yu; Y. Andrew Wang; Yanqiang Zhong; Amanda R. Welton; Craig Galbán; Shaomeng Wang; Duxin Sun

Studies were conducted to develop antibody- and fluorescence-labeled superparamagnetic iron oxide nanoparticle (SPIO) nanotheranostics for magnetic resonance imaging (MRI) and fluorescence imaging of cancer cells and pH-dependent intracellular drug release. SPIO nanoparticles (10 nm) were coated with amphiphilic polymers and PEGylated. The antibody HuCC49ΔCH2 and fluorescent dye 5-FAM were conjugated to the PEG of iron oxide nanoparticles (IONPs). Anticancer drugs doxorubicin (Dox), azido-doxorubicin (Adox), MI-219, and 17-DMAG containing primary amine, azide, secondary amine, and tertiary amine, respectively, were encapsulated into IONPs. The encapsulation efficiency and drug release at various pHs were determined using LC-MS/MS. The cancer targeting and imaging were monitored using MRI and fluorescent microscopy in a colon cancer cell line (LS174T). The pH-dependent drug release, intracellular distribution, and cytotoxicity were evaluated using microscopy and MTS assay. The PEGylation of SPIO and conjugation with antibody and 5-FAM increased SPIO size from 18 to 44 nm. Fluorescent imaging, magnetic resonance imaging (MRI) and Prussian blue staining demonstrated that HuCC49ΔCH2-SPIO increased cancer cell targeting. HuCC49ΔCH2-SPIO nanotheranostics decreased the T(2) values in MRI of LS174T cells from 117.3 ± 1.8 ms to 55.5 ± 2.6 ms. The loading capacities of Dox, Adox, MI-219, and 17-DMAG were 3.16 ± 0.77%, 6.04 ± 0.61%, 2.22 ± 0.42%, and 0.09 ± 0.07%, respectively. Dox, MI-219 and 17-DMAG showed pH-dependent release while Adox did not. Fluorescent imaging demonstrated the accumulation of HuCC49ΔCH2-SPIO nanotheranostics in endosomes/lysosomes. The encapsulated Dox was released in acidic lysosomes and diffused into cytosol and nuclei. In contrast, the encapsulated Adox only showed limited release in endosomes/lysosomes. HuCC49ΔCH2-SPIO nanotheranostics target-delivered more Dox to LS174T cells than nonspecific IgG-SPIO and resulted in a lower IC(50) (1.44 μM vs 0.44 μM). The developed HuCC49ΔCH2-SPIO nanotheranostics provides an integrated platform for cancer cell imaging, targeted anticancer drug delivery and pH-dependently drug release.


Aaps Journal | 2012

Applications of Human Pharmacokinetic Prediction in First-in-Human Dose Estimation

Peng Zou; Yanke Yu; Nan Zheng; Yongsheng Yang; Hayley J. Paholak; Lawrence X. Yu; Duxin Sun

Quantitative estimations of first-in-human (FIH) doses are critical for phase I clinical trials in drug development. Human pharmacokinetic (PK) prediction methods have been developed to project the human clearance (CL) and bioavailability with reasonable accuracy, which facilitates estimation of a safe yet efficacious FIH dose. However, the FIH dose estimation is still very challenging and complex. The aim of this article is to review the common approaches for FIH dose estimation with an emphasis on PK-guided estimation. We discuss 5 methods for FIH dose estimation, 17 approaches for the prediction of human CL, 6 methods for the prediction of bioavailability, and 3 tools for the prediction of PK profiles. This review may serve as a practical protocol for PK- or pharmacokinetic/pharmacodynamic-guided estimation of the FIH dose.


Journal of Nutritional Biochemistry | 2012

Sulforaphane inhibits pancreatic cancer through disrupting Hsp90-p50Cdc37 complex and direct interactions with amino acids residues of Hsp90

Yanyan Li; G. Elif Karagöz; Young Ho Seo; Tao Zhang; Yiqun Jiang; Yanke Yu; Afonso M.S. Duarte; Steven J. Schwartz; Rolf Boelens; Kate S. Carroll; Stefan Rüdiger; Duxin Sun

Sulforaphane [1-isothiocyanato-4-(methyl-sulfinyl) butane)], an isothiocyanate derived from cruciferous vegetables, has been shown to possess potent chemopreventive activity. We analyzed the effect of sulforaphane on the proliferation of pancreatic cancer cells. Sulforaphane inhibited pancreatic cancer cell growth in vitro with IC(50)s of around 10-15 μM and induced apoptosis. In pancreatic cancer xenograft mouse model, administration of sulforaphane showed remarkable inhibition of tumor growth without apparent toxicity noticed. We found that sulforaphane induced the degradation of heat shock protein 90 (Hsp90) client proteins and blocked the interaction of Hsp90 with its cochaperone p50(Cdc37) in pancreatic cancer cells. Using nuclear magnetic resonance spectroscopy (NMR) with an isoleucine-specific labeling strategy, we overcame the protein size limit of conventional NMR and studied the interaction of sulforaphane with full-length Hsp90 dimer (170 kDa) in solution. NMR revealed multiple chemical shifts in sheet 2 and the adjacent loop in Hsp90 N-terminal domain after incubation of Hsp90 with sulforaphane. Liquid chromatography coupled to mass spectrometry further mapped a short peptide in this region that was tagged with sulforaphane. These data suggest a new mechanism of sulforaphane that disrupts protein-protein interaction in Hsp90 complex for its chemopreventive activity.


Journal of Biological Chemistry | 2010

Split Renilla Luciferase Protein Fragment-assisted Complementation (SRL-PFAC) to Characterize Hsp90-Cdc37 Complex and Identify Critical Residues in Protein/Protein Interactions

Yiqun Jiang; Denzil Bernard; Yanke Yu; Yehua Xie; Tao Zhang; Yanyan Li; Joseph Burnett; Xueqi Fu; Shaomeng Wang; Duxin Sun

Hsp90 requires cochaperone Cdc37 to load its clients to the Hsp90 superchaperone complex. The purpose of this study was to utilize split Renilla luciferase protein fragment-assisted complementation (SRL-PFAC) bioluminescence to study the full-length human Hsp90-Cdc37 complex and to identity critical residues and their contributions for Hsp90/Cdc37 interaction in living cells. SRL-PFAC showed that full-length human Hsp90/Cdc37 interaction restored dramatically high luciferase activity through Hsp90-Cdc37-assisted complementation of the N and C termini of luciferase (compared with the set of controls). Immunoprecipitation confirmed that the expressed fusion proteins (NRL-Hsp90 and Cdc37-CRL) preserved their ability to interact with each other and also with native Hsp90 or Cdc37. Molecular dynamic simulation revealed several critical residues in the two interaction patches (hydrophobic and polar) at the interface of Hsp90/Cdc37. Mutagenesis confirmed the critical residues for Hsp90-Cdc37 complex formation. SRL-PFAC bioluminescence evaluated the contributions of these critical residues in Hsp90/Cdc37 interaction. The results showed that mutations in Hsp90 (Q133A, F134A, and A121N) and mutations in Cdc37 (M164A, R167A, L205A, and Q208A) reduced the Hsp90/Cdc37 interaction by 70–95% as measured by the resorted luciferase activity through Hsp90-Cdc37-assisted complementation. In comparison, mutations in Hsp90 (E47A and S113A) and a mutation in Cdc37 (A204E) decreased the Hsp90/Cdc37 interaction by 50%. In contrast, mutations of Hsp90 (R46A, S50A, C481A, and C598A) and mutations in Cdc37 (C54S, C57S, and C64S) did not change Hsp90/Cdc37 interactions. The data suggest that single amino acid mutation in the interface of Hsp90/Cdc37 is sufficient to disrupt its interaction, although Hsp90/Cdc37 interactions are through large regions of hydrophobic and polar interactions. These findings provides a rationale to develop inhibitors for disruption of the Hsp90/Cdc37 interaction.


Journal of Pharmacy and Pharmaceutical Sciences | 2012

Preclinical Pharmacokinetics of MI-219, a Novel Human Double Minute 2 (HDM2) Inhibitor and Prediction of Human Pharmacokinetics

Peng Zou; Nan Zheng; Yanke Yu; Shanghai Yu; Wei Sun; Donna McEachem; Yongsheng Yang; Lawrence X. Yu; Shaomeg Wang; Duxin Sun

PURPOSE The two purposes of this study were evaluating preclinical pharmacokinetics of MI-219 and predicting clearance (CL) and volume of distribution at steady-state (Vdss) of MI-219 in humans. METHODS Pharmacokinetic studies were conducted on mice, rats, dogs, and monkeys. Human CL of MI-219 was predicted using allometric scaling (SA), multi-exponential allometric scaling (ME), rule of exponents (RoE), single species scaling, two-term power equation (TTPE), physiologically based in vitro-in vivo extrapolation (IVIVE), and fu corrected intercept method (FCIM). In vitro assays were conducted to determine in vitro intrinsic CL, protein binding, and blood-plasma partition coefficients. To estimate half-life of MI-219, plasma concentration-time profile in humans was predicted using kallynochron and apolysichron time transformation (Dedrick plots) and normalization with MRT and Vdss (Wajimas method). In addition, simultaneous interspecies scaling of CL, Vdss and concentration-time profile were performed by using Nonlinear Mixed Effects Modeling (NONMEM). RESULTS Preclinical studies showed that the elimination of MI-219 was mainly through metabolism. The validation using observed monkey CL and Vdss showed that MA, IVIVE and Oie-Tozer methods were accurately than the other methods. Human CL of MI-219 predicted by ME and IVIVE was between 0.237-0.342 L*h⁻¹*kg⁻¹. Human Vdss predicted by Oie-Tozer method and allometric scaling of unbound volume of distribution of tissues (VT/fuT) method was between 0.93-1.40 L*kg⁻¹. Superimposition of rat, monkey and dog data was observed in Dedrick plots and Wajimas transformations. CONCLUSIONS The predicted human pharmacokinetics is useful for the design of first-in-human study.


Cancer Research | 2010

Abstract 3392: Heat shock protein 27 increases metastasis of human head and neck squamous cancer cells in vitro

Zhenkun Zhu; Xin Xu; Yanke Yu; Martin P. Graham; Gangli Liu; Mark E. Prince; Thomas E. Carey; Duxin Sun

The small heat shock protein 27 (Hsp27) is a molecular chaperone that is involved in a variety of cellular functions in cancer cells. The purpose of this research is to study the function of Hsp27 in metastasis of head and neck squamous cancer cells. The expression of Hsp27 in primary and metastatic head and neck cancer cells was determined using real-time PCR and western blotting. The siRNA knockdown of Hsp27 was performed in metastatic head and neck cancer cells using in vitro transfection. The proliferation of the primary and metastatic head and neck cancer cells were evaluated using MTS proliferation assay. The metastatic potential was determined using both migration assay and invasion assay in metastatic head and neck cancer cells after siRNA silencing of Hsp27. MTS proliferation showed that both primary (UM-SCC-22A) and metastatic (UM-SCC-22B) head and neck cancer cells have similar proliferation rate from 24-48 hrs. However, the metastatic head and neck cancer cells (UM-SCC-22B) showed more than 2-fold migration ability and more than 3-fold invasion ability than primary head and neck cancer cells (UM-SCC-22A). Real-time PCR demonstrated that Hsp27 mRNA is 22-fold higher in metastatic UM-SCC-22B than primary UM-SCC-22A. Similarly, Western blotting exhibited that primary UM-SCC-22A did not show any detectable levels of Hsp27, while metastatic UM-SCC-22B showed significant high levels (> 40-fold) of Hsp27. siRNA of Hsp27 knocked down expression of Hsp27 by more than 5-fold in mRNA level and 23-fold in protein levels in metastatic head and neck cancer cells UM-SCC-22B. Furthermore, siRNA knockdown of Hsp27 decreased metastasis of UM-SCC-22B by more than 50% in cell invasion assay. These data suggest that Hsp27 may regulate metastatic potential of head and neck squamous cancer cells. Silencing Hsp27 may decrease metastatic potential of head and neck squamous cancer cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3392.


Cancer Research | 2010

Abstract 2672: Characterization of Hsp90/Cdc37 interaction and their critical residues using luciferase fragment complementation imaging

Yiqun Jiang; Denzil Bernard; Yanke Yu; Tao Zhang; Yanyan Li; Xueqi Fu; Shaomeng Wang; Duxin Sun

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC The purpose of this study is to investigate Hsp90/Cdc37 interaction and identify residues critical for the interaction using real-time luciferase complementation imaging and computational modeling. Molecular imaging using a split Renilla luciferase fragment complementation (SRL-FC) was established to monitor real-time Hsp90/Cdc37interactions in living cells. Computational modeling and molecular dynamics simulations were used to evaluate the interaction interface of Hsp90/Cdc37 complex and critical residues in the interaction interface were identified for mutagenesis. Western Blotting and co-immunoprecipitation were used to determine protein expression and complex formation. The molecular imaging in living cells showed that Hsp90 and Cdc37 helped the complementation of N-terminus of Renilla luciferase (fused with Hsp90; N-RL-Hsp90) and C-terminus of Renilla luciferase (fused with Cdc37, Cdc37-C-RL) with 170-fold higher luciferase activity than controls without Hsp90/Cdc37. This suggests that the SRL-FC is sensitive and specific to monitor the interaction of full length human Hsp90 and Cdc37. Molecular dynamics simulations revealed that the interface was constituted by a series of residues interacting through hydrophobic or polar interactions. Mutagenesis confirmed that mutations in Hsp90 (A121N, Q133A, F134A) and mutations in Cdc37 (M164K, R167A) led to the loss of complementation of N-RL-Hsp90 and Cdc37-C-RL (5 to 10-fold lower luciferase activity compared to wild-type), indicating these residues are critical in Hsp90/Cdc37 interactions. In comparison, mutations in Hsp90 (E47A, S113A) and mutations in Cdc37 (A204E) only decreased 50% of the complementation of N-RL-Hsp90 and Cdc37-C-RL, which suggests that these residues contribute less to Hsp90/Cdc37 interaction. In contrast, mutations of Hsp90 (R46A, S50A) and mutation in Cdc37 (L165H) did not change the complementation of N-RL-Hsp90 and Cdc37-C-RL, indicating that these residues did not contribute to Hsp90/Cdc37 interaction. The data suggests that molecular imaging using split Renilla luciferase fragment complementation (SRL-FC) can be used to identify the critical residues in Hsp90/Cdc37 interaction. Although Hsp90/Cdc37 interactions are through both hydrophobic and polar interactions, mutation in a single amino acid residue, including Ala121, Gln133, Phe134 in human Hsp90 and Met164, Arg167 in human Cdc37, is sufficient to disrupt Hsp90/Cdc37 interaction. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2672.

Collaboration


Dive into the Yanke Yu's collaboration.

Top Co-Authors

Avatar

Duxin Sun

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Tao Zhang

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Yanyan Li

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Peng Zou

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Yiqun Jiang

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge