Network


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

Hotspot


Dive into the research topics where Dunyong Tan is active.

Publication


Featured researches published by Dunyong Tan.


Journal of Molecular Endocrinology | 2010

Short form 1b human prolactin receptor down-regulates expression of the long form

Dunyong Tan; Ameae M. Walker

Alternative splicing produces different human prolactin (PRL) receptors. These include a long form (LF) and two short forms (SF1a and SF1b). The SFs of the receptor can act as dominant negatives for PRL effector function through the LF. This is proposed to be due to LF-SF heterodimerization and resultant interference with LF-LF dimer signaling. We, along with others, have provided evidence for LF-SF heterodimerization of the human receptors in support of this mechanism, along with others. However, to further investigate the ways SF may influence LF function, we co-transfected human embryonic kidney 293 cells with vectors coding for tagged (green fluorescent protein (GFP) or luciferase) LF alone or plus untagged SF1b and measured LF-GFP intensity, LF-luciferase activity, and LF mRNA 48 h later. Equal amounts of SF1b cDNA decreased LF-GFP fluorescence intensity, LF-luciferase activity, and LF mRNA by 80-100%. Similar co-transfections with untagged LF had no significant effect on tagged LF expression. Use of hygromycin showed degradation of already formed protein was the same for LF-luciferase alone and LF-luciferase with SF1b. Inhibition of mRNA synthesis, on the other hand, showed that SF1b expression accelerated LF mRNA degradation two- to three-fold. SF1b also down-regulated expression of endogenous LF mRNA in T47D breast cancer cells and opposed an increase in cell number resulting from transfection with extra LF alone. These results demonstrate a previously unrecognized mechanism whereby SF1b affects the end result of signaling through the LF receptor. The effects on cell number also support the concept that the LF:SF1b ratio may be relevant to tumor growth.


Cancer Letters | 2011

Prolactin increases survival and migration of ovarian cancer cells: Importance of prolactin receptor type and therapeutic potential of S179D and G129R receptor antagonists

Dunyong Tan; KuanHui E. Chen; Teresa Khoo; Ameae M. Walker

Variably-spliced prolactin receptors (PRLRs) and PRL are expressed by the ovarian cancer cell lines, TOV-112D, OV-90 and TOV-21G. Incubation in the PRLR antagonists, G129R- or S179D-PRL, or anti-PRL reduced cell number, indicating a functional autocrine PRL growth loop. Added PRL promoted, and the antagonists decreased, cell migration. When cells were stressed, added PRL decreased apoptosis and increased survival, and the antagonists had the opposite effect. Cells expressing higher long:short PRLR ratios had increased growth, survival and migration in response to PRL. Results suggest that PRLR antagonists may be therapeutically beneficial in ovarian cancer.


Oncology Research | 2006

Different forms of prolactin have opposing effects on the expression of cell cycle regulatory proteins in differentiated mammary epithelial cells.

Wei Wu; Yen Hao Chen; Eric Ueda; Dunyong Tan; Paolo Bartolini; Ameae M. Walker

Prolactin (PRL) is a hormone that contributes to both the growth and differentiation of mammary epithelial cells, activities likely to impact breast cancer in opposite ways. Whether PRL causes growth or differentiation has been solely attributed to the coexisting steroidal environment, with PRL stimulating mammary gland growth during pregnancy, and then milk production after the postpartum drop in estrogen and progesterone. However, previous work from our laboratory has shown that the form of PRL may also be an important factor. During pregnancy, unmodified PRL (U-PRL) promotes mammary growth, while an increase in phosphorylated PRL, or administration of a molecular mimic of phosphorylated PRL (S179D PRL), inhibits growth. Unknown, however, is whether these forms of PRL have opposite effects on growth in the absence of steroids and whether effects are directly on mammary epithelial cells. To mimic the glandular epithelium in vitro, we used contact-inhibited, differentiated cells and showed that even with these minimally growing cells that treatment with U-PRL caused increased expression of cyclin D1 and cyclin-dependent kinase 4, increased activity of both cdk4 and cdk2, while having no effect on the inhibitory protein, p21. S179D PRL, by contrast, had no effect on cyclin D1 and cdk4 expression, but increased p21 expression and expression of the vitamin D receptor (VDR). We conclude that increased U-PRL or decreased phosphorylated PRL can directly affect cell cycle control proteins in relatively differentiated mammary epithelial cells, thereby implicating the balance between these two forms of PRL in the early promotion of breast cancer.


Chinese Journal of Physiology | 2013

Histone Trimethylation of the p53 Gene by Expression of a Constitutively Active Prolactin Receptor in Prostate Cancer Cells

Dunyong Tan; Si Tan; Jie Zhang; Peizhi Tang; Jian-Jun Huang; Wei-Hua Zhou; Shi-Jun Wu

Prolactin (PRL) is a pituitary polypeptide hormone characterized by multiple biological actions including stimulation of growth in the prostate, breast and ovarian epithelial cells. A sizable body of reports has presented evidences to indicate the involvement of PRL in the pathogenic process of cancers of the reproductive system, such as prostate and breast cancers. PRL exerts its effects by dimerizing its receptor (PRLR) on the plasma membrane, and initiating cellular Jak-Stat signal pathway. We have previously cloned from prostate cancer cells a natural variant of PRLR in which the S2 subdomain of the extracellular domain is missing (ΔS2). Our preliminary data showed that ΔS2 PRLR was able to dimerize and to constitutively activate the β-casein promoter (in the absence of its ligand, PRL) in breast and prostate epithelial cells. Enhancer of zeste homologue 2 (EZH2), an important histone-modifying enzyme, is able to trimethylate histone 3 on lysine 27 (H3K27Me3), consequently leading to gene silencing, especially silencing of tumor suppressor genes such as p53. We hypothesized that ΔS2 PRLR played an important pathogenic role in prostate cancer through, at least partly, alterations in the expression of EZH2 and the trimethylation of histone 3 on lysine 27. In the present study, overexpression of ΔS2 PRLR in prostate epithelial cells was achieved by infection with an adenoviral vector carrying the cDNA. The viable cell number overexpressing ΔS2 PRLR was assessed using MTS reagent. Western blot, chromatin immunoprecipitation (ChIP) assay and acid histone extraction were applied to detect expression of EZH2 as well as trimethylation of histone 3, respectively. In prostate epithelial cells, overexpression of ΔS2 PRLR increased the levels of EZH2 methyltransferase mRNA and protein, induced EZH2 methyltransferase recruitment to chromatin, increased the trimethylation of histone 3 on lysine 27, and decreased expression of the p53 gene. We concluded that ΔS2 PRLR plays an important pathogenic role in prostate cancer through epigenetic covalent modification leading to chromatin remodeling. Hypertrimethylation on H3K27 of the p53 gene promoter region due to elevated expression of ΔS2 PRLR by alternative splicing of the pre-mRNA in its full-length form might serve as a new mechanism underlying human prostate cancer.


Cancer Letters | 2014

An N-terminal splice variant of human Stat5a that interacts with different transcription factors is the dominant form expressed in invasive ductal carcinoma.

Dunyong Tan; KuanHui E. Chen; Changhui Deng; Peizhi Tang; Jian-Jun Huang; Trina Mansour; Richard A. Luben; Ameae M. Walker

We have identified a new variant of human Stat5a, found at higher ratios to full-length Stat5a in invasive ductal carcinoma versus contiguous normal tissue. The variant, missing exon 5, inhibits p21 and Bax production and increases cell number. After prolactin stimulation, only full-length Stat5a interacts with the vitamin D and retinoid X receptors, whereas only Δ5 Stat5a interacts with activating protein 1-2 and specificity protein 1. Prolactin also oppositely regulates interaction of the two Stat5a forms with β-catenin. We propose that a change in splicing leading to upregulation of this new isoform is a pathogenic aspect of invasive ductal carcinoma.


Cancer Letters | 2015

Blockade of estrogen-stimulated proliferation by a constitutively-active prolactin receptor having lower expression in invasive ductal carcinoma.

KuangTzu Huang; Dunyong Tan; KuanHui E. Chen; Ameae M. Walker

A comprehensive understanding of prolactins (PRLs) role in breast cancer is complicated by disparate roles for alternatively-spliced PRL receptors (PRLR) and crosstalk between PRL and estrogen signaling. Among PRLRs, the short form 1b (SF1b) inhibits PRL-stimulated cell proliferation. In addition to ligand-dependent PRLRs, constitutively-active varieties, missing the S2 region of the extracellular domain (ΔS2), naturally occur. Expression analysis of the ΔS2 version of SF1b (ΔS2SF1b) showed higher expression in histologically-normal contiguous tissue versus invasive ductal carcinoma. To determine the function of ΔS2SF1b, a T47D breast cancer line with inducible expression was produced. Induction of ΔS2SF1b blocked estrogen-stimulated cell proliferation. Unlike intact SF1b, induction of ΔS2SF1b had no effect on PRL-mediated activation of Stat5a. However induction inhibited estrogens stimulatory effects on serine-118 phosphorylation of estrogen receptor α, serine-473 phosphorylation of Akt, serine-9 phosphorylation of GSK3β, and c-myc expression. In addition, induction of ΔS2SF1b increased expression of the cell cycle-inhibiting protein, p21. Thus, increased expression of ΔS2SF1b, such as we demonstrate occurs with the selective PRLR modulator, S179D PRL, would create a physiological state in which estrogen-stimulated proliferation was inhibited, but differentiative responses to PRL were maintained.


Adaptive Medicine | 2012

Association of Sodium Induced High Blood Pressure with Up-Regulation of Inducible Nitric Oxide Synthase (iNOS) Expression and Inhibition of Trimethylation of H3K27 on Its Promoter Region

Dunyong Tan; Si Tan; Jie Zhang; Wei-Hua Zhou; Shi-Jun Wu

Nitric oxide (NO), a simple molecule, playing very diverse roles in the regulation of the cardiovascular, immunologic and nervous system, is formed from L-arginine by converting it to L-citrulline via nitric oxide synthase enzymes (NOS) including endothelial NOS (eNOS), neuronal NOS (nNOS) and inducible NOS (iNOS). We previously reported that long term inhibition of iNOS by intravenous administration of aminogunidine (AG) led to an aggravation of hypertension induced by high sodium chloride (NaCl) loading. Also, high blood pressure induced by high NaCl loading is associated with higher NO production in SD rats. The goal of the present study was to clarify whether the change of blood pressure alter iNOS gene expression as well as epigenetic modification of iNOS gene promoter region. Up-regulation of iNOS mRNA and protein were observed in renal medullary tissue of high sodium infused rats. Furthermore, chromatin immunoprecipitation assay evidenced the inhibition of recruitment of methyltransferase enhancer of Zeste homology 2 (EZH2) to, and trimethylation of histone 3 (H3) at lysine 27 (K27) of iNOS gene promoter region. We concluded that blood pressure is a regulator of iNOS gene expression through alteration of histone covalent modification of its genomic DNA.


Chinese Journal of Physiology | 2015

Histone 3 Trimethylation of IGFBP-7 Gene Promoter by Expression of D5 Stat5a in Breast Epithelial Cells

Dunyong Tan; Jie Zhang; Xi Nie; Chao Yu; Zhong-Zheng Li; Zulbnur Mamat; Huang Jiang; Xiao-Meng Yang; Jiao Zhang; Sihong Yao; Peizhi Tang; Jian-Jun Huang

As a very important transcription factor, signal transducer and activator of transcription 5a (Stat5a) has been reported to be involved in human reproductive cancers such as breast, prostate and ovarian cancer. However, up to date, the exact role of Stat5a in breast cancer is still not clear. The data reported are conflicting. D5 Stat5a is a variant of Stat5a we cloned recently. This newly cloned variant behave like its full length counterpart in terms of dimerization, being activated by prolactin and nuclear translocation, however it also behave differently in terms of effect on cell proliferation and interaction with other transcription factors. In the present study, we examined its effect on cell proliferation of cultured breast cancer cells (MCF-10A and MCF-7) by using adenovirus-mediated gene transfer and MTS technology. Also, quantitative real time polymerase chain reaction (qRT-PCR), chromatin immunoprecipitation assay (ChIP) and Western blot were used to probe the possible changes of insulin-like growth factor binding protein-7 (IGFBP-7) expression including mRNA and protein, and the epigenetic changes with overexpression of this newly cloned variant. The results clarified that D5 Stat5a (1) behaves as a promoting factor to the cell proliferation of MCF-10A and MCF-7, (2) induces enhancer of zeste homology 2 (EZH2) expression in breast epithelial cells, as well as histone 3 trimethylation of IGFBP-7 promoter region, and (3) lower IGFBP-7 expression was detected in breast cancer tissue. Taking together, we concluded that the mitogenic effect of D5 Stat5a on breast cells is, at least partly, through up-regulation of histone methyltransferase, EZH2, and therefore inhibiting IGFBP-7 expression by increasing H3K27Me3 of IGFBP-7 promoter region.


Adaptive Medicine | 2013

Association of D5 Stat5a Induced Breast Cancer Cell Migration with Increased H3K27 Trimethylation and Down-Regulated Expression of E-Cadherin Gene

Dunyong Tan; Jie Zhang; Peizhi Tang; Jian-Jun Huang; Ameae M. Walker

As a very important transcription factor, signal transducer and activator of transcription 5a (Stat5a) has been reported to be involved in human reproductive cancers such as breast, prostate and ovarian cancer. However, up to date, the exact role of Stat5a in breast cancer is still not clear. The data reported are conflicting. D5 Stat5a is a variant of Stat5a we cloned previously. In the present study, we examined its effect on the migration of cultured breast cancer cells (MCF-7) by using adenovirus-mediated gene transfer and transwell technology. Also, chromatin immunoprecipitation (ChIP) assay and quantitative real time polymerase chain reaction (qRT-PCR) were applied to probe to epigenetic changes with overexpression of this newly cloned variant. The results showed that D5 Stat5a behave very differently from its full length counterpart, Stat5a, in cell migration of breast cancer cells. Stat5a inhibits, but D5 Stat5a promotes the migration of breast cancer cells. ChIP and qRT-PCR evidenced that overexpression of this variant increased trimethylation of lysine 27 on histone 3 (H3K27Me3) of E-Cadherin promoter region. The new form of Stat5a and the results of the present study may account for, at least partly, the conflict phenomenon reported by different investigators regarding the role of Stat5a in reproductive cancer.


Molecular Endocrinology | 2005

Unmodified Prolactin (PRL) and S179D PRL-Initiated Bioluminescence Resonance Energy Transfer between Homo- and Hetero-Pairs of Long and Short Human PRL Receptors in Living Human Cells

Dunyong Tan; David A. Johnson; Wei Wu; Lingfang Zeng; Yen Hao Chen; Wen Y. Chen; Barbara K. Vonderhaar; Ameae M. Walker

Collaboration


Dive into the Dunyong Tan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wei Wu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yen Hao Chen

University of California

View shared research outputs
Top Co-Authors

Avatar

Eric Ueda

University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Barbara K. Vonderhaar

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Changhui Deng

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge