Network


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

Hotspot


Dive into the research topics where Yunguang Tong is active.

Publication


Featured researches published by Yunguang Tong.


Oncogene | 2007

Pituitary tumor transforming gene interacts with Sp1 to modulate G1/S cell phase transition

Yunguang Tong; Y Tan; Cuiqi Zhou; Shlomo Melmed

Pituitary tumor transforming gene (PTTG1) was isolated from rat pituitary tumor cells, and subsequently identified as a securin protein as well as a transcription factor. We show here a global transcriptional effect of PTTG1 in human choriocarcinoma JEG-3 cells by simultaneously assessing 20 000 gene promoters using chromatin immunoprecipitation (ChIP)-on-Chip experiments. Seven hundred and forty-six gene promoters (P<0.001) were found enriched, with functions relating to cell cycle, metabolic control and signal transduction. Significant interaction between PTTG1 and Sp1 (P<0.000001) was found by transcriptional pattern analysis of ChIP data and further confirmed by immunoprecipitation and pull-down assays. PTTG1 acts coordinately with Sp1 to induce cyclin D3 expression ∼threefold, and promotes G1/S-phase transition independently of p21. PTTG1 deletion was also protective for anchorage-independent cell colony formation. The results show that PTTG1 exhibits properties of a global transcription factor, and specifically modulates the G1/S-phase transition by interacting with Sp1. This novel signaling pathway may be required for PTTG1 transforming activity.


PLOS ONE | 2011

Lineage-Specific Restraint of Pituitary Gonadotroph Cell Adenoma Growth

Vera Chesnokova; Svetlana Zonis; Cuiqi Zhou; Anat Ben-Shlomo; Kolja Wawrowsky; Yoel Toledano; Yunguang Tong; Kalman Kovacs; Bernd W. Scheithauer; Shlomo Melmed

Although pituitary adenomas are usually benign, unique trophic mechanisms restraining cell proliferation are unclear. As GH-secreting adenomas are associated with p53/p21-dependent senescence, we tested mechanisms constraining non-functioning pituitary adenoma growth. Thirty six gonadotroph-derived non-functioning pituitary adenomas all exhibited DNA damage, but undetectable p21 expression. However, these adenomas all expressed p16, and >90% abundantly expressed cytoplasmic clusterin associated with induction of the Cdk inhibitor p15 in 70% of gonadotroph and in 26% of somatotroph lineage adenomas (p = 0.006). Murine LβT2 and αT3 gonadotroph pituitary cells, and αGSU.PTTG transgenic mice with targeted gonadotroph cell adenomas also abundantly expressed clusterin and exhibited features of oncogene-induced senescence as evidenced by C/EBPβ and C/EBPδ induction. In turn, C/EBPs activated the clusterin promoter ∼5 fold, and elevated clusterin subsequently elicited p15 and p16 expression, acting to arrest murine gonadotroph cell proliferation. In contrast, specific clusterin suppression by RNAis enhanced gonadotroph proliferation. FOXL2, a tissue-specific gonadotroph lineage factor, also induced the clusterin promoter ∼3 fold in αT3 pituitary cells. As nine of 12 pituitary carcinomas were devoid of clusterin expression, this protein may limit proliferation of benign adenomatous pituitary cells. These results point to lineage-specific pathways restricting uncontrolled murine and human pituitary gonadotroph adenoma cell growth.


Oncogene | 2014

PTTG acts as a STAT3 target gene for colorectal cancer cell growth and motility

Cuiqi Zhou; Yunguang Tong; Kolja Wawrowsky; Shlomo Melmed

Pituitary tumor-transforming gene (PTTG), the index mammalian securin, is abundantly expressed in several tumors and regulates tumor growth and progression. Molecular mechanisms elucidating PTTG regulation and actions remain elusive. Here, we provide evidence that PTTG acts as a signal transducer and activator of transcription factor 3 (STAT3) target gene. Total STAT3 and Tyr705 phosphorylated STAT3 were concordantly expressed with PTTG in human colorectal tumors (n=97 and n=95, respectively, P<0.001). STAT3 specifically bound the human PTTG promoter and induced PTTG transcriptional activity (twofold) as assessed by chromatin immunoprecipitation and luciferase reporter assays. STAT3 transfection increased PTTG mRNA and protein abundance twofold in HCT116 human colon cancer cells, and induction was further enhanced (threefold) by constitutively active STAT3 (STAT3-C), whereas strongly abrogated by dominant-negative STAT3 (STAT3-DN). Attenuating PTTG expression by siRNA in STAT3 HCT116 stable transfectants suppressed cell growth and colony formation in vitro, and PTTG cell knockout also constrained activated STAT3-induced explanted murine tumor growth in vivo. STAT3 increased HCT116 cell migration and invasion up to fivefold, whereas cell mobility was abolished by STAT3-DN (>85%). Impairing PTTG expression by siRNA also strongly suppressed STAT3-faciliated cell migration and invasion by up to 90%. Knocking out PTTG in STAT3-C HCT116 stable transfectants strongly decreased tumor metastases in nude mice, indicating the requirement of PTTG for STAT3-promoted metastasis. These results elucidate a mechanism for tumor cell PTTG regulation, whereby STAT3 induces PTTG expression to facilitate tumor growth and metastasis, and further support the rationale for targeting PTTG to abrogate colorectal cancer growth.


Oncogene | 2008

Pituitary tumor transforming gene 1 regulates Aurora kinase A activity.

Yunguang Tong; A Ben-Shlomo; Cuiqi Zhou; Kolja Wawrowsky; Shlomo Melmed

Pituitary tumor transforming gene 1 (PTTG1), a transforming gene highly expressed in several cancers, is a mammalian securin protein regulating both G1/S and G2/M phases. Using protein array screening, we showed PTTG1 interacting with Aurora kinase A (Aurora-A), and confirmed the interaction using co-immunoprecipitation, His-tagged pull-down assays and intracellular immunofluorescence colocalization. PTTG1 transfection into HCT116 cells prevented Aurora-A T288 autophosphorylation, inhibited phosphorylation of the histone H3 Aurora-A substrate and resulted in abnormally condensed chromatin. PTTG1-null cell proliferation was more sensitive to Aurora-A knock down and to Aurora kinase Inhibitor III treatment. The results indicate that PTTG1 and Aurora-A interact to regulate cellular responses to anti-neoplastic drugs. PTTG1 knockdown is therefore a potential approach to improve the efficacy of tumor Aurora kinase inhibitors.


Chinese Journal of Cancer | 2011

Mouse models of colorectal cancer

Yunguang Tong; Wancai Yang; H. Phillip Koeffler

Colorectal cancer is one of the most common malignancies in the world. Many mouse models have been developed to evaluate features of colorectal cancer in humans. These can be grouped into genetically-engineered, chemically-induced, and inoculated models. However, none recapitulates all of the characteristics of human colorectal cancer. It is critical to use a specific mouse model to address a particular research question. Here, we review commonly used mouse models for human colorectal cancer.


Endocrinology | 2012

Genomic Characterization of Human and Rat Prolactinomas

Yunguang Tong; Yun Zheng; Jin Zhou; Nelson M. Oyesiku; H. Phillip Koeffler; Shlomo Melmed

Although prolactinomas can be effectively treated with dopamine agonists, about 20% of patients develop dopamine resistance or tumor recurrence after surgery, indicating a need for better understanding of underlying disease mechanisms. Although estrogen-induced rat prolactinomas have been widely used to investigate the development of this tumor, the extent that the model recapitulates features of human prolactinomas is unclear. To prioritize candidate genes and gene sets regulating human and rat prolactinomas, microarray results derived from human prolactinomas and pituitaries of estrogen-treated ACI rats were integrated and analyzed. A total of 4545 differentially expressed pituitary genes were identified in estrogen-treated ACI rats [false discovery rate (FDR) < 0.01]. By comparing pituitary microarray results derived from estrogen-treated Brown Norway rats (a strain not sensitive to estrogen), 4073 genes were shown specific to estrogen-treated ACI rats. Human prolactinomas exhibited 1177 differentially expressed genes (FDR < 0.05). Combining microarray data derived from human prolactinoma and pituitaries of estrogen-treated ACI rat, 145 concordantly expressed genes, including E2F1, Myc, Igf1, and CEBPD, were identified. Gene set enrichment analysis revealed that 278 curated pathways and 59 gene sets of transcription factors were enriched (FDR < 25%) in estrogen-treated ACI rats, suggesting a critical role for Myc, E2F1, CEBPD, and Sp1 in this rat prolactinoma. Similarly increased Myc, E2F1, and Sp1 expression was validated using real-time PCR and Western blot in estrogen-treated Fischer rat pituitary glands. In summary, characterization of individual genes and gene sets in human and in estrogen-induced rat prolactinomas validates the model and provides insights into genomic changes associated with this commonly encountered pituitary tumor.


Molecular Endocrinology | 2011

CEBPD Suppresses Prolactin Expression and Prolactinoma Cell Proliferation

Yunguang Tong; Jin Zhou; Jun Mizutani; Hidenori Fukuoka; Song Guang Ren; Arthur Gutierrez-Hartmann; H. Phillip Koeffler; Shlomo Melmed

Hyperprolactinemia, usually caused by a pituitary lactotroph tumor, leads to galactorrhea and infertility. Increased prolactin (PRL) levels may be due to enhanced PRL expression or proliferation of PRL-secreting cells. We hypothesize that PRL expression and PRL-secreting cell proliferation are linked. Using microarray-based gene expression profiling, we identified CCAAT-enhancer-binding protein δ (CEBPD) transcription factor as a critical gene that regulates both PRL expression and lactotroph cell proliferation. CEBPD expression levels are decreased approximately 7-fold in experimental rat prolactinoma cells. Forced expression of this transcription factor in PRL-secreting cells (GH3 and MMQ) inhibited PRL expression and cellular proliferation, and CEBPD knockdown by small interfering RNA leads to increased PRL expression in both cell lines. To determine mechanisms underlying this observation, we determined binding of CEBPD to the PRL promoter and also showed marked suppression (96%) of PRL promoter activity. CEBPD and Pit1 interact and attenuate each others binding to the PRL promoter. CEBPD also suppresses expression of proliferation-related genes, including c-Myc, survivin, as well as cyclins B1, B2, and D1. These results show that PRL expression and cell proliferation are controlled in part by CEBPD.


PLOS ONE | 2011

PTTG1 Attenuates Drug-Induced Cellular Senescence

Yunguang Tong; Weijiang Zhao; Cuiqi Zhou; Kolja Wawrowsky; Shlomo Melmed

As PTTG1 (pituitary tumor transforming gene) abundance correlates with adverse outcomes in cancer treatment, we determined mechanisms underlying this observation by assessing the role of PTTG1 in regulating cell response to anti-neoplastic drugs. HCT116 cells devoid of PTTG1 (PTTG1−/−) exhibited enhanced drug sensitivity as assessed by measuring BrdU incorporation in vitro. Apoptosis, mitosis catastrophe or DNA damage were not detected, but features of senescence were observed using low doses of doxorubicin and TSA. The number of drug-induced PTTG1−/− senescent cells increased ∼4 fold as compared to WT PTTG1-replete cells (p<0.001). p21, an important regulator of cell senescence, was induced ∼3 fold in HCT116 PTTG1−/− cells upon doxorubicin or Trichostatin A treatment. Binding of Sp1, p53 and p300 to the p21 promoter was enhanced in PTTG1−/− cells after treatment, suggesting transcriptional regulation of p21. p21 knock down abrogated the observed senescent effects of these drugs, indicating that PTTG1 likely suppresses p21 to regulate drug-induced senescence. PTTG1 also regulated SW620 colon cancer cells response to doxorubicin and TSA mediated by p21. Subcutaneously xenografted PTTG1−/− HCT116 cells developed smaller tumors and exhibited enhanced responses to doxorubicin. PTTG1−/− tumor tissue derived from excised tumors exhibited increased doxorubicin-induced senescence. As senescence is a determinant of cell responses to anti-neoplastic treatments, these findings suggest PTTG1 as a tumor cell marker to predict anti-neoplastic treatment outcomes.


Endocrine-related Cancer | 2008

Oct-1 induces pituitary tumor transforming gene expression in endocrine tumors

Cuiqi Zhou; Yunguang Tong; Kolja Wawrowsky; Serguei Bannykh; Ines Donangelo; Shlomo Melmed

As human pituitary tumor transforming gene (hPTTG1) is upregulated in endocrine tumors, we studied regulatory mechanisms for hPTTG1 expression. We identified Oct-1-binding motifs in the hPTTG1 promoter region and show Oct-1-specific binding to the hPTTG1 promoter using chromatin immunoprecipitation. We overexpressed Oct-1 and observed approximately 2.5-fold activation of hPTTG1 promoter luciferase constructs (-2642/-1 and -1717/-1). Transcriptional activation was abrogated by co-transfection of an inactive Oct-1 form lacking the POU domain or by utilizing mutated hPTTG1 promoters or mutants devoid of two Oct-1-binding motifs (-1717/-1mut, -637/-1 or -433/-1). Using biotin-streptavidin pull-down assays, we confirmed Oct-1 binding to the two octamer motifs in the hPTTG1 promoter (-1669/-1631 and -1401/-1361). Endogenous hPTTG1 mRNA and protein increased up to approximately fourfold in Oct-1 transfectants, as measured by real-time PCR and western blot. In contrast, siRNA-mediated suppression of endogenous Oct-1 attenuated both the hPTTG1 mRNA and protein levels. Using confocal immunofluorescence imaging, Oct-1 and hPTTG1 were concordantly upregulated in pituitary (57 and 62%, n=79, P<0.01) and breast tumor specimens (57 and 42%, n=77, P<0.05) respectively. The results show that Oct-1 transactivates hPTTG1, and both proteins are concordantly overexpressed in endocrine tumors, thus offering a mechanism for endocrine tumor hPTTG1 abundance.


Cancer Research | 2014

Abstract 2081: FoxM1 transactivates PTTG1 and promotes colorectal cancer cell migration and invasion

Jinjun Guo; Yun Zheng; Yunguang Tong; H. Philip Koeffler

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Previous studies have shown that both Forkhead Box M1 (FoxM1) and pituitary tumor transforming gene 1 (PTTG1) were associated with cancer development and metastases. This study aims to investigate the relation between FoxM1 and PTTG1 and its role in colorectal cancer cell behavior. The microarray data of colorectal cancer were analyzed to determine the association of FoxM1 and PTTG1 expression. Luciferase reporter assay, ChIP and EMSA were used to identify the regulatory mechanisms between FoxM1 and PTTG1. PTTG1 mRNA and protein levels were examined using real-time PCR and western blot after knocking down or over expressing FoxM1. The roles of FoxM1 and PTTG in tumor metastasis were investigated by cell migration and invasion assays. Gene expression microarray data indicated that FoxM1 and PTTG1 are concordantly up-regulated in colorectal cancer tissues. Over-expression of FoxM1 up-regulated PTTG1 expression and knock-down of FoxM1 did the opposite, indicating that PTTG1 was regulated by FoxM1. Luciferase reporter assay indicated that -391 to +50 bps of PTTG1 promoter are important for FoxM1 to enhance PTTG1 promoter activity. Sequence analysis identified two potential FoxM1 binding sites. EMSA indicated that FoxM1 directly binds to PTTG1 promoter at the -350 to -343 binding motif. This binding was confirmed by chromatin immunoprecipitation in HCT116 and SW620 cells. Boyden chamber assay indicated that FoxM1 and PTTG1 concordantly regulate migration and invasion of HCT116 and SW620 colorectal cancer cells. In summary, our results elucidate that FoxM1 up-regulates PTTG1 transcription through binding to PTTG1 promoter. PTTG1 likely mediates effects of FoxM1 on colorectal cancer cell migration and invasion. Citation Format: Jinjun Guo, Yun Zheng, Yunguang Tong, H Philip Koeffler. FoxM1 transactivates PTTG1 and promotes colorectal cancer cell migration and invasion. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2081. doi:10.1158/1538-7445.AM2014-2081

Collaboration


Dive into the Yunguang Tong's collaboration.

Top Co-Authors

Avatar

Shlomo Melmed

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cuiqi Zhou

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kolja Wawrowsky

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Serguei Bannykh

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Beth Y. Karlan

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dong-Joo Cheon

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jin Zhou

University of Arizona

View shared research outputs
Top Co-Authors

Avatar

Jun Mizutani

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mourad Tighiouart

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sandra Orsulic

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge