Network


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

Hotspot


Dive into the research topics where Yiyu Dong is active.

Publication


Featured researches published by Yiyu Dong.


Science Signaling | 2013

PUMA and BIM Are Required for Oncogene Inactivation–Induced Apoptosis

Gregory R. Bean; Yogesh Tengarai Ganesan; Yiyu Dong; Shugaku Takeda; Han Liu; Po M. Chan; Yafen Huang; Lewis A. Chodosh; Gerard P. Zambetti; James J. Hsieh; Emily H. Cheng

Combining drugs that inhibit antiapoptotic proteins with oncogenic kinase inhibitors may overcome drug resistance in cancer therapy. Using Addiction Against Cancer Oncogene-addicted cancer cells depend on a particular oncogenic protein for survival and die when the oncogenic protein is inactivated. For example, lung cancers with an abnormally active form of the epidermal growth factor receptor (EGFR) and breast cancers with amplification of the gene encoding the human epidermal growth factor receptor 2 (HER2) regress when exposed to drugs called tyrosine kinase inhibitors that block the activity of these receptors. Bean et al. analyzed breast and lung cancer cell lines treated with tyrosine kinase inhibitors and mice in which EGFRs were inactivated by genetic ablation. They found that two signaling pathways, the phosphoinositide 3-kinase (PI3K)–AKT and mitogen-activated or extracellular signal–regulated protein kinase kinase (MEK)–extracellular signal–regulated kinase (ERK) pathways, were inactivated, resulting in an increase in the abundance of PUMA and BIM, two proteins that promote cell death. Furthermore, cancer cell lines resistant to tyrosine kinase inhibitors were killed by the combination of a PI3K inhibitor and a drug that inhibits antiapoptotic proteins. Thus, pharmacologically enhancing the death pathway may overcome or prevent resistance to tyrosine kinase inhibitors. The clinical efficacy of tyrosine kinase inhibitors supports the dependence of distinct subsets of cancers on specific driver mutations for survival, a phenomenon called “oncogene addiction.” We demonstrate that PUMA and BIM are the key apoptotic effectors of tyrosine kinase inhibitors in breast cancers with amplification of the gene encoding human epidermal growth factor receptor 2 (HER2) and lung cancers with epidermal growth factor receptor (EGFR) mutants. The BH3 domain containing proteins BIM and PUMA can directly activate the proapoptotic proteins BAX and BAK to permeabilize mitochondria, leading to caspase activation and apoptosis. We delineated the signal transduction pathways leading to the induction of BIM and PUMA by tyrosine kinase inhibitors. Inhibition of the mitogen-activated or extracellular signal–regulated protein kinase kinase (MEK)–extracellular signal–regulated kinase (ERK) pathway caused increased abundance of BIM, whereas antagonizing the phosphoinositide 3-kinase (PI3K)–AKT pathway triggered nuclear translocation of the FOXO transcription factors, which directly activated the PUMA promoter. In a mouse breast tumor model, the abundance of PUMA and BIM was increased after inactivation of HER2. Moreover, deficiency of Bim or Puma impaired caspase activation and reduced tumor regression caused by inactivation of HER2. Similarly, deficiency of Puma impeded the regression of EGFRL858R-driven mouse lung tumors upon inactivation of the EGFR-activating mutant. Overall, our study identified PUMA and BIM as the sentinels that interconnect kinase signaling networks and the mitochondrion-dependent apoptotic program, which offers therapeutic insights for designing novel cell death mechanism–based anticancer strategies.


Journal of Clinical Investigation | 2013

HGF-MET signals via the MLL-ETS2 complex in hepatocellular carcinoma

Shugaku Takeda; Han Liu; Satoru Sasagawa; Yiyu Dong; Paul A. Trainor; Emily H. Cheng; James J. Hsieh

HGF signals through its cognate receptor, MET, to orchestrate diverse biological processes, including cell proliferation, cell fate specification, organogenesis, and epithelial-mesenchymal transition. Mixed-lineage leukemia (MLL), an epigenetic regulator, plays critical roles in cell fate, stem cell, and cell cycle decisions. Here, we describe a role for MLL in the HGF-MET signaling pathway. We found a shared phenotype among Mll(-/-), Hgf(-/-), and Met(-/-) mice with common cranial nerve XII (CNXII) outgrowth and myoblast migration defects. Phenotypic analysis demonstrated that MLL was required for HGF-induced invasion and metastatic growth of hepatocellular carcinoma cell lines. HGF-MET signaling resulted in the accumulation of ETS2, which interacted with MLL to transactivate MMP1 and MMP3. ChIP assays demonstrated that activation of the HGF-MET pathway resulted in increased occupancy of the MLL-ETS2 complex on MMP1 and MMP3 promoters, where MLL trimethylated histone H3 lysine 4 (H3K4), activating transcription. Our results present an epigenetic link between MLL and the HGF-MET signaling pathway, which may suggest new strategies for therapeutic intervention.


Cell Reports | 2017

The SWI/SNF protein PBRM1 restrains VHL-loss-driven clear cell renal cell carcinoma

Amrita M. Nargund; Can G. Pham; Yiyu Dong; Patricia Wang; Hatice U. Osmangeyoglu; Yuchen Xie; Omer Aras; Song Han; Toshinao Oyama; Shugaku Takeda; Chelsea E. Ray; Zhenghong Dong; Mathieu Berge; A. Ari Hakimi; Sebastien Monette; Carl L. Lekaye; Jason A. Koutcher; Christina S. Leslie; Chad J. Creighton; Nils Weinhold; William R. Lee; Satish K. Tickoo; Zhong Wang; Emily H. Cheng; James J. Hsieh

PBRM1 is the second most commonly mutated gene after VHL in clear cell renal cell carcinoma (ccRCC). However, the biological consequences of PBRM1 mutations for kidney tumorigenesis are unknown. Here, we find that kidney-specific deletion of Vhl and Pbrm1, but not either gene alone, results in bilateral, multifocal, transplantable clear cell kidney cancers. PBRM1 loss amplified the transcriptional outputs of HIF1 and STAT3 incurred by Vhl deficiency. Analysis of mouse and human ccRCC revealed convergence on mTOR activation, representing the third driver event after genetic inactivation of VHL and PBRM1. Our study reports a physiological preclinical ccRCC mouse model that recapitulates somatic mutations in human ccRCC and provides mechanistic and therapeutic insights into PBRM1 mutated subtypes of human ccRCC.


Journal of Clinical Investigation | 2016

Mechanistically distinct cancer-associated mTOR activation clusters predict sensitivity to rapamycin

Jianing Xu; Can G. Pham; Steven K. Albanese; Yiyu Dong; Toshinao Oyama; Chung-Han Lee; Vanessa Rodrik-Outmezguine; Zhan Yao; Song Han; David Y. T. Chen; Daniel L. Parton; John D. Chodera; Neal Rosen; Emily H. Cheng; James J. Hsieh

Genomic studies have linked mTORC1 pathway-activating mutations with exceptional response to treatment with allosteric inhibitors of mTORC1 called rapalogs. Rapalogs are approved for selected cancer types, including kidney and breast cancers. Here, we used sequencing data from 22 human kidney cancer cases to identify the activating mechanisms conferred by mTOR mutations observed in human cancers and advance precision therapeutics. mTOR mutations that clustered in focal adhesion kinase targeting domain (FAT) and kinase domains enhanced mTORC1 kinase activity, decreased nutrient reliance, and increased cell size. We identified 3 distinct mechanisms of hyperactivation, including reduced binding to DEP domain-containing MTOR-interacting protein (DEPTOR), resistance to regulatory associated protein of mTOR-mediated (RAPTOR-mediated) suppression, and altered kinase kinetics. Of the 28 mTOR double mutants, activating mutations could be divided into 6 complementation groups, resulting in synergistic Rag- and Ras homolog enriched in brain-independent (RHEB-independent) mTORC1 activation. mTOR mutants were resistant to DNA damage-inducible transcript 1-mediated (REDD1-mediated) inhibition, confirming that activating mutations can bypass the negative feedback pathway formed between HIF1 and mTORC1 in the absence of von Hippel-Lindau (VHL) tumor suppressor expression. Moreover, VHL-deficient cells that expressed activating mTOR mutants grew tumors that were sensitive to rapamycin treatment. These data may explain the high incidence of mTOR mutations observed in clear cell kidney cancer, where VHL loss and HIF activation is pathognomonic. Our study provides mechanistic and therapeutic insights concerning mTOR mutations in human diseases.


Cancer Research | 2012

A pharmacologic inhibitor of the protease Taspase1 effectively inhibits breast and brain tumor growth.

David Y. T. Chen; Yishan Lee; Brian A. Van Tine; Adam C. Searleman; Todd D. Westergard; Han Liu; Ho Chou Tu; Shugaku Takeda; Yiyu Dong; David Piwnica-Worms; Kyoung J. Oh; Stanley J. Korsmeyer; Ann Hermone; Richard Gussio; Robert H. Shoemaker; Emily H. Cheng; James J. Hsieh

The threonine endopeptidase Taspase1 has a critical role in cancer cell proliferation and apoptosis. In this study, we developed and evaluated small molecule inhibitors of Taspase1 as a new candidate class of therapeutic modalities. Genetic deletion of Taspase1 in the mouse produced no overt deficiencies, suggesting the possibility of a wide therapeutic index for use of Taspase1 inhibitors in cancers. We defined the peptidyl motifs recognized by Taspase1 and conducted a cell-based dual-fluorescent proteolytic screen of the National Cancer Institute diversity library to identify Taspase1 inhibitors (TASPIN). On the basis of secondary and tertiary screens the 4-[(4-arsonophenyl)methyl]phenyl] arsonic acid NSC48300 was determined to be the most specific active compound. Structure-activity relationship studies indicated a crucial role for the arsenic acid moiety in mediating Taspase1 inhibition. Additional fluorescence resonance energy transfer-based kinetic analysis characterized NSC48300 as a reversible, noncompetitive inhibitor of Taspase1 (K(i) = 4.22 μmol/L). In the MMTV-neu mouse model of breast cancer and the U251 xenograft model of brain cancer, NSC48300 produced effective tumor growth inhibition. Our results offer an initial preclinical proof-of-concept to develop TASPINs for cancer therapy.


Nature Communications | 2016

Molecular analysis of aggressive renal cell carcinoma with unclassified histology reveals distinct subsets.

Ying Bei Chen; Jianing Xu; Anders Skanderup; Yiyu Dong; A. Rose Brannon; Lu Wang; Helen H. Won; Patricia Wang; Gouri Nanjangud; Achim A. Jungbluth; Wei Li; Virginia Ojeda; A. Ari Hakimi; Martin H. Voss; Nikolaus Schultz; Robert J. Motzer; Paul Russo; Emily H. Cheng; Filippo G. Giancotti; William R. Lee; Michael F. Berger; Satish K. Tickoo; Victor E. Reuter; James J. Hsieh

Renal cell carcinomas with unclassified histology (uRCC) constitute a significant portion of aggressive non-clear cell renal cell carcinomas that have no standard therapy. The oncogenic drivers in these tumours are unknown. Here we perform a molecular analysis of 62 high-grade primary uRCC, incorporating targeted cancer gene sequencing, RNA sequencing, single-nucleotide polymorphism array, fluorescence in situ hybridization, immunohistochemistry and cell-based assays. We identify recurrent somatic mutations in 29 genes, including NF2 (18%), SETD2 (18%), BAP1 (13%), KMT2C (10%) and MTOR (8%). Integrated analysis reveals a subset of 26% uRCC characterized by NF2 loss, dysregulated Hippo–YAP pathway and worse survival, whereas 21% uRCC with mutations of MTOR, TSC1, TSC2 or PTEN and hyperactive mTORC1 signalling are associated with better clinical outcome. FH deficiency (6%), chromatin/DNA damage regulator mutations (21%) and ALK translocation (2%) distinguish additional cases. Altogether, this study reveals distinct molecular subsets for 76% of our uRCC cohort, which could have diagnostic and therapeutic implications.


Nature Communications | 2017

Targeting the differential addiction to anti-apoptotic BCL-2 family for cancer therapy

Akane Inoue-Yamauchi; Paul S Jeng; Kwanghee Kim; Hui-Chen Chen; Song Han; Yogesh Tengarai Ganesan; Kota Ishizawa; Sylvia Jebiwott; Yiyu Dong; Maria Catherine Pietanza; Matthew D. Hellmann; Mark G. Kris; James J. Hsieh; Emily H. Cheng

BCL-2 family proteins are central regulators of mitochondrial apoptosis and validated anti-cancer targets. Using small cell lung cancer (SCLC) as a model, we demonstrated the presence of differential addiction of cancer cells to anti-apoptotic BCL-2, BCL-XL or MCL-1, which correlated with the respective protein expression ratio. ABT-263 (navitoclax), a BCL-2/BCL-XL inhibitor, prevented BCL-XL from sequestering activator BH3-only molecules (BH3s) and BAX but not BAK. Consequently, ABT-263 failed to kill BCL-XL-addicted cells with low activator BH3s and BCL-XL overabundance conferred resistance to ABT-263. High-throughput screening identified anthracyclines including doxorubicin and CDK9 inhibitors including dinaciclib that synergized with ABT-263 through downregulation of MCL-1. As doxorubicin and dinaciclib also reduced BCL-XL, the combinations of BCL-2 inhibitor ABT-199 (venetoclax) with doxorubicin or dinaciclib provided effective therapeutic strategies for SCLC. Altogether, our study highlights the need for mechanism-guided targeting of anti-apoptotic BCL-2 proteins to effectively activate the mitochondrial cell death programme to kill cancer cells.


Cell Research | 2014

Taspase1 cleaves MLL1 to activate cyclin E for HER2/neu breast tumorigenesis.

Yiyu Dong; Brian A. Van Tine; Toshinao Oyama; Patricia Wang; Emily H. Cheng; James J. Hsieh

Taspase1, a highly conserved threonine protease, cleaves nuclear transcriptional regulators mixed-lineage leukemia (MLL, MLL1), MLL2, TFIIA, and ALF to orchestrate a wide variety of biological processes. In vitro studies thus far demonstrated that Taspase1 plays important roles in the proliferation of various cancer cell lines, including HER2-positive breast cancer cells. To investigate the role of Taspase1 in breast tumorigenesis in vivo, we deleted Taspase1 from mouse mammary glands by generating MMTV-neu;MMTV-cre;Tasp1F/− mice. We demonstrate that initiation of MMTV-neu- but not MMTV-wnt-driven breast cancer is blocked in the absence of Taspase1. Importantly, Taspase1 loss alone neither impacts normal development nor pregnancy physiology of the mammary gland. In mammary glands Taspase1 deficiency abrogates MMTV-neu-induced cyclins E and A expression, thereby preventing tumorigenesis. The mechanisms were explored in HER2-positive breast cancer cell line BT474 and HER2-transformed MCF10A cells and validated using knockdown-resistant Taspase1. As Taspase1 was shown to cleave MLL which forms complexes with E2F transcription factors to regulate Cyclins E, A, and B expression in mouse embryonic fibroblasts (MEFs), we investigated whether the cleavage of MLL by Taspase1 constitutes an essential in vivo axis for HER2/neu-induced mammary tumorigenesis. To this end, we generated MMTV-neu;MLLnc/nc transgenic mice that carry homozygous non-cleavable MLL alleles. Remarkably, these mice are also protected from HER2/neu-driven breast tumorigenesis. Hence, MLL is the primary Taspase1 substrate whose cleavage is required for MMTV-neu-induced tumor formation. As Taspase1 plays critical roles in breast cancer pathology, it may serve as a therapeutic target for HER2-positive human breast cancer.


international conference of the ieee engineering in medicine and biology society | 2015

Mathematical models of tumor growth using Voronoi tessellations in pathology slides of kidney cancer

Aydin Saribudak; Yiyu Dong; Stephen Gundry; James J. Hsieh; M. Ümit Uyar

The impact of patient-specific spatial distribution features of cell nuclei on tumor growth characteristics was analyzed. Tumor tissues from kidney cancer patients were allowed to grow in mice to apply H&E staining and to measure tumor volume during preclinical phase of our study. Imaging the H&E stained slides under a digital light microscope, the morphological characteristics of nuclei positions were determined. Using artificial intelligence based techniques, Voronoi features were derived from diagrams, where cell nuclei were considered as distinct nodes. By identifying the effect of each Voronoi feature, tumor growth was expressed mathematically. Consistency between the computed growth curves and preclinical measurements indicates that the information obtained from the H&E slides can be used as biomarkers to build personalized mathematical models for tumor growth.


Nature Communications | 2017

Analysis of renal cancer cell lines from two major resources enables genomics-guided cell line selection

Rileen Sinha; Andrew G. Winer; Michael Chevinsky; Christopher Jakubowski; Ying-Bei Chen; Yiyu Dong; Satish K. Tickoo; Victor E. Reuter; Paul Russo; Jonathan A. Coleman; Chris Sander; James J. Hsieh; A. Ari Hakimi

The utility of cancer cell lines is affected by the similarity to endogenous tumour cells. Here we compare genomic data from 65 kidney-derived cell lines from the Cancer Cell Line Encyclopedia and the COSMIC Cell Lines Project to three renal cancer subtypes from The Cancer Genome Atlas: clear cell renal cell carcinoma (ccRCC, also known as kidney renal clear cell carcinoma), papillary (pRCC, also known as kidney papillary) and chromophobe (chRCC, also known as kidney chromophobe) renal cell carcinoma. Clustering copy number alterations shows that most cell lines resemble ccRCC, a few (including some often used as models of ccRCC) resemble pRCC, and none resemble chRCC. Human ccRCC tumours clustering with cell lines display clinical and genomic features of more aggressive disease, suggesting that cell lines best represent aggressive tumours. We stratify mutations and copy number alterations for important kidney cancer genes by the consistency between databases, and classify cell lines into established gene expression-based indolent and aggressive subtypes. Our results could aid investigators in analysing appropriate renal cancer cell lines.

Collaboration


Dive into the Yiyu Dong's collaboration.

Top Co-Authors

Avatar

James J. Hsieh

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Emily H. Cheng

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

A. Ari Hakimi

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jonathan A. Coleman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Song Han

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Paul Russo

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shugaku Takeda

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Han Liu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kwanghee Kim

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sylvia Jebiwott

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge