Network


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

Hotspot


Dive into the research topics where Heetae Kim is active.

Publication


Featured researches published by Heetae Kim.


Oncogene | 2008

The AP-1 transcription factor regulates breast cancer cell growth via cyclins and E2F factors

Qiang Shen; Ivan P. Uray; Yi Li; Tibor Krisko; Tracy Strecker; Heetae Kim; Powel H. Brown

The activating protein-1 (AP-1) transcription factor transduces growth signals through signal transduction pathways to the nucleus, leading to the expression of genes involved in growth and malignant transformation in many cell types. We have previously shown that overexpression of a dominant negative form of the cJun proto-oncogene, a cJun dominant negative mutant (Tam67), blocks AP-1 transcriptional activity, induces a G1 cell cycle block and inhibits breast cancer cell growth in vitro and in vivo. We found that AP-1 blockade by Tam67 in MCF-7 breast cancer cells downregulates cyclin D1 transcriptional activity by at least two mechanisms: by suppressing transcription at the known AP-1 binding site (−934/−928) and by suppressing growth factor-induced expression through suppressing E2F activation at the E2F-responsive site (−726/−719). AP-1 blockade also led to reduced expression of E2F1 and E2F2, but not E2F4, at the mRNA and protein levels. Chromatin immunoprecipitation and supershift assays demonstrated that AP-1 blockade caused decreased binding of E2F1 protein to the E2F site in the cyclin D1 promoter. We also found that Tam67 suppressed the expression of the E2F1 dimerizing partner, DP1 and E2F-upregulated cell cycle genes (cyclins E, A, B and D3) and enhanced the expression of E2F-downregulated cell cycle genes (cyclins G2 and I). Reduced expression of other E2F-regulated genes was also seen with AP-1 blockade and E2F suppression. Thus, the AP-1 factor regulates the expression of cyclin D and E2F (the latter in turn regulates E2F-downstream genes), leading to cell cycle progression and breast cancer cell proliferation.


Journal of Biological Chemistry | 2006

Mitogen-activated Protein Kinase Kinase-4 Promotes Cell Survival by Decreasing PTEN Expression through an NFκB-dependent Pathway

Dianren Xia; Harish Srinivas; Young Ho Ahn; Gautam Sethi; Xiaoyang Sheng; W. K. Alfred Yung; Qianghua Xia; Paul J. Chiao; Heetae Kim; Powel H. Brown; Ignacio I. Wistuba; Bharat B. Aggarwal; Jonathan M. Kurie

Mitogen-activated protein kinase kinase-4 (MKK4/SEK1) cooperates with phosphatidylinositol 3-kinase to maintain the survival of non-small cell lung cancer (NSCLC) cells, but the biochemical basis of this phenomenon has not been elucidated. Here we used genetic approaches to modulate MKK4 expression in mouse embryo fibroblasts (MEF cells) and NSCLC cells to identify prosurvival signals downstream of MKK4. Relative to wild-type MEF cells, MKK4-null MEF cells were highly susceptible to apoptosis by LY294002, paclitaxel, or serum starvation. MKK4 promoted the survival of MEF cells by decreasing the expression of phosphatase and tensin homologue deleted from chromosome 10 (PTEN). MKK4 inhibited PTEN transcription by activating NFκB, a transcriptional suppressor of PTEN. MKK4 was required for nuclear translocation of RelA/p65 and processing of the NFκB2 precursor (p100) into the mature form (p52). Studies on a panel of NSCLC cell lines revealed a subset with high MKK4/high NFκB/low PTEN that was relatively resistant to apoptosis. Thus, MKK4 promotes cell survival by activating phosphatidylinositol 3-kinase through an NFκB/PTEN-dependent pathway.


Molecular Endocrinology | 2011

Estrogen Induces c-myc Gene Expression via an Upstream Enhancer Activated by the Estrogen Receptor and the AP-1 Transcription Factor

Chunyu Wang; Julie Ann Mayer; Abhijit Mazumdar; Kirsten Fertuck; Heetae Kim; Myles Brown; Powel H. Brown

c-myc oncogene is implicated in tumorigenesis of many cancers, including breast cancer. Although c-myc is a well-known estrogen-induced gene, its promoter has no estrogen-response element, and the underlying mechanism by which estrogen induces its expression remains obscure. Recent genome-wide studies by us and others suggested that distant elements may mediate estrogen induction of gene expression. In this study, we investigated the molecular mechanism by which estrogen induces c-myc expression with a focus on these distal elements. Estrogen rapidly induced c-myc expression in estrogen receptor (ER)-positive breast cancer cells. Although estrogen had little effect on c-myc proximal promoter activity, it did stimulate the activity of a luciferase reporter containing a distal 67-kb enhancer. Estrogen induction of this luciferase reporter was dependent upon both a half-estrogen response element and an activator protein 1 (AP-1) site within this enhancer, which are conserved across 11 different mammalian species. Small interfering RNA experiments and chromatin immunoprecipitation assays demonstrated the necessity of ER and AP-1 cross talk for estrogen to induce c-myc expression. TAM67, the AP-1 dominant negative, partially inhibited estrogen induction of c-myc expression and suppressed estrogen-induced cell cycle progression. Together, these results demonstrate a novel pathway of estrogen regulation of gene expression by cooperation between ER and AP-1 at the distal enhancer element and that AP-1 is involved in estrogen induction of the c-myc oncogene. These results solve the long-standing question in the field of endocrinology of how estrogen induces c-myc expression.


Oncogene | 2004

AP-1 blockade in breast cancer cells causes cell cycle arrest by suppressing G1 cyclin expression and reducing cyclin-dependent kinase activity

Yongmin Liu; Chunhua Lu; Qiang Shen; Debbie Munoz-Medellin; Heetae Kim; Powel H. Brown

The AP-1 transcription factor is a central component of signal transduction pathways in many cells, although the exact role of AP-1 in controlling cell growth and malignant transformation is unknown. We have previously shown that AP-1 complexes are activated by peptide and steroid growth factors in both normal and malignant breast cells, and that blocking AP-1 by overexpressing a dominant-negative form of cJun (cJun-DN, TAM67) inhibits breast cancer cell growth both in vivo and in vitro. We hypothesized that TAM67 inhibits cell growth by altering the expression of cell cycle regulatory proteins, thus causing a cell cycle block. In the present study, we used clones of MCF7 breast cancer cells that express TAM67 under the control of an inducible promoter. First, we determined the effect of AP-1 blockade on cell growth, then we performed 3H-thymidine incorporation and flow cytometry assays to investigate whether TAM67 inhibits the cell cycle. We observed that in the presence of serum TAM67 inhibited cell growth and caused a block in the G1 phase of the cell cycle. Next, we performed Western-blotting and CDK kinase assays to determine the effects of TAM67 on retinoblastoma (Rb) phosphorylation, the expression of cell cycle regulatory proteins, and CDK activity. We discovered that TAM67 inhibited Rb phosphorylation and reduced E2F activity. We also found that TAM67 decreased the expression of D and E cyclins, reduced CDK2 and CDK4 activity, and increased the CDK inhibitor p27. The studies of gene expression at the RNA level showed that TAM67 decreased cyclin Ds mRNA expression. Our study suggests that in the presence of serum, TAM67 inhibits breast cancer growth predominantly by inducing inhibitors of cyclin-dependent kinases (such as p27) and by reducing the expression of the cyclins involved in transitioning from G1 into S phase of the cell cycle. These studies lay the foundation for future attempt to develop new agents for the treatment and prevention of breast cancer.


Oncogene | 2005

cFos is critical for MCF-7 breast cancer cell growth

Chunhua Lu; Qiang Shen; Elizabeth DuPré; Heetae Kim; Susan G. Hilsenbeck; Powel H. Brown

The activating protein-1 (AP-1) transcription factor is a converging point of multiple signal transduction pathways in many cells. We have previously demonstrated that overexpressing Tam67, a dominant-negative (DN) form of cJun, blocks AP-1 activity and inhibits breast cancer cell growth. We hypothesized that Tam67 forms dimers with other AP-1 proteins to suppress the growth of breast cancer cells. In the present study, we used immunoprecipitation-Western blotting to demonstrate that Tam67 binds all Jun and Fos proteins in breast cancer cells. In addition, we used two variants of the Tam67 mutant to investigate whether Jun or Fos protein was required for breast cancer cell growth. We created a Tam/Fos mutant in which the cJun dimerization domain was replaced by the cFos dimerization domain, and a Tam/Squelcher mutant in which the cJun dimerization domain was deleted. We then isolated MCF-7 cell lines that stably expressed these cJun-DN mutants under the control of an inducible promoter. Using AP-1-dependent reporter assays, we observed that Tam67 and Tam/Fos mutants inhibited AP-1 transcriptional activity, while the Tam/Squelcher mutant did not. We then determined whether Tam/Fos or Tam/Squelcher inhibited breast cell growth as well as Tam67. We found that while Tam67 repressed cell growth, neither Tam/Fos nor Tam/Squelcher mutant affected cell growth. These results indicate that Tam67 likely inactivates Fos family member proteins to suppress breast cancer cell growth. Finally, we performed antisense experiments to knock down the expression of individual family members (cJun or cFos). Our results demonstrated that antisense cFos inhibited breast cancer cell proliferation and colony formation, while antisense cJun did not. These results suggest that Tam67 suppresses breast cancer cell growth by interacting with Fos family members, specifically with cFos, to produce an inactive AP-1 complex.


Clinical Cancer Research | 2006

The combination of the rexinoid, LG100268, and a selective estrogen receptor modulator, either arzoxifene or acolbifene, synergizes in the prevention and treatment of mammary tumors in an estrogen receptor-negative model of breast cancer

Karen T. Liby; Mara H. Rendi; Nanjoo Suh; Darlene B. Royce; Renee Risingsong; Charlotte R. Williams; William W. Lamph; Fernand Labrie; Stan Krajewski; Xiao Chun Xu; Heetae Kim; Powel H. Brown; Michael B. Sporn

Purpose: We tested whether a selective estrogen receptor modulator (SERM) and a rexinoid are active for prevention and treatment in the mouse mammary tumor virus-neu mouse model of estrogen receptor–negative breast cancer. Experimental Design: For prevention, mice were fed a powdered control diet, the SERM arzoxifene (Arz, 20 mg/kg diet), the rexinoid LG100268 (268, 30 mg/kg diet), or the combination for 60 weeks. In a second prevention study, mice were fed Arz (6 mg/kg diet), 268 (30 mg/kg diet), the combination of Arz and 268, the SERM acolbifene (Acol, 3 mg/kg diet), or the combination of Acol and 268 for 52 weeks. For the treatment studies, mice with tumors were fed combinations of a SERM and 268 for 4 weeks. Results: The rexinoid 268 and the SERMs Arz and Acol, as individual drugs, delayed the development of estrogen receptor–negative tumors. Moreover, the combination of a SERM and 268 was strikingly synergistic, as no tumors developed in any mouse fed the combination of 268 and a SERM. Moreover, this drug combination also induced significant tumor regression when used therapeutically. These drugs did not inhibit transgene expression in vitro or in vivo, and the combination of Arz and 268 inhibited proliferation and induced apoptosis in the tumors. Conclusion: The combination of a rexinoid and SERM should be considered for future clinical trials.


British Journal of Cancer | 2008

The rexinoid, bexarotene, prevents the development of premalignant lesions in MMTV-erbB2 mice

Yi Li; Yun Zhang; Jamal Hill; Heetae Kim; Qiang Shen; Reid P. Bissonnette; William W. Lamph; Powel H Brown

Retinoids, vitamin A analogues that bind to retinoic acid receptor (RAR) or retinoid X receptor (RXR), play important roles in regulating cell proliferation, apoptosis, and differentiation. Recently, RXR-selective ligands, also referred to as rexinoids, have been investigated as potential chemopreventive agents for breast cancer. Our previous studies demonstrated that the rexinoid bexarotene significantly prevented ER-negative mammary tumourigenesis with less toxicity than naturally occurring retinoids in animal models. To determine whether bexarotene prevents cancer at the early stages during the multistage process of mammary carcinogenesis, we treated MMTV-erbB2 mice with bexarotene for 2 or 4 months. The development of preinvasive mammary lesions such as hyperplasias and carcinoma-in-situ was significantly inhibited. This inhibition was associated with reduced proliferation, but no induction of apoptosis. We also examined the regulation of a number of rexinoid-modulated genes including critical growth and cell cycle regulating genes using breast cell lines and mammary gland samples from mice treated with rexinoids. We showed that two of these genes (DHRS3 and DEC2) were modulated by bexarotene both in vitro and in vivo. Identification of these rexinoid-modulated genes will help us understand the mechanism by which rexinoid prevents cancer. Such rexinoid-regulated genes also represent potential biomarkers to assess the response of rexinoid treatment in clinical trials.


Biochemical Journal | 2006

Akt phosphorylates and suppresses the transactivation of retinoic acid receptor α

Harish Srinivas; Dianren Xia; Nicole L. Moore; Ivan P. Uray; Heetae Kim; Long Ma; Nancy L. Weigel; Powel H. Brown; Jonathan M. Kurie

The transactivation of nuclear receptors is regulated by both ligand binding and phosphorylation. We previously showed that RARalpha (retinoic acid receptor alpha) phosphorylation by c-Jun N-terminal kinase contributes to retinoid resistance in a subset of NSCLC cells (non-small cell lung cancer cells), but the aetiology of this resistance in the remainder has not been fully elucidated [Srinivas, Juroske, Kalyankrishna, Cody, Price, Xu, Narayanan, Weigel and Kurie (2005) Mol. Cell. Biol. 25, 1054-1069]. In the present study, we report that Akt, which is constitutively activated in NSCLC cells, phosphorylates RARalpha and inhibits its transactivation. Biochemical and functional analyses showed that Akt interacts with RARalpha and phosphorylates the Ser96 residue of its DNA-binding domain. Mutation of Ser96 to alanine abrogated the suppressive effect of Akt. Overexpression of a dominant-negative form of Akt in an NSCLC cell line decreased RAR phosphorylation, increased RAR transactivation and enhanced the growth-inhibitory effects of an RAR ligand. The findings presented here show that Akt inhibits RAR transactivation and contributes to retinoid resistance in a subset of NSCLC cells.


Breast Cancer Research and Treatment | 2006

Receptor-selective retinoids inhibit the growth of normal and malignant breast cells by inducing G1 cell cycle blockade

Kendall Wu; Elizabeth DuPré; Heetae Kim; Caesar K. Tin-U; Reid P. Bissonnette; William W. Lamph; Powel H. Brown

SummaryDespite advances in treatment, breast cancer continues to be the second leading cause of cancer mortality in women. Statistics suggest that while focus on treatment should continue, chemopreventive approaches should also be pursued. Previous studies have demonstrated that naturally occurring retinoids such as 9-cis retinoic acid (9cRA) can prevent breast cancer in animal models. However, these studies have also shown that these compounds are too toxic for general use. Work from our laboratory showed that an RXR-selective retinoid LGD1069 prevented tumor development in animal models of cancer with reduced toxicity as compared to an RAR-selective retinoid TTNPB. In the present study, we investigated the mechanisms by which receptor-selective retinoids inhibit the growth of normal and malignant breast cells. Our results demonstrate that the synthetic retinoids tested are as effective as 9cRA in suppressing the growth of normal human mammary epithelial cells (HMECs) and estrogen receptor-positive (ER-positive) breast cancer cells. Although the receptor-selective retinoids induce minimal amounts of apoptosis in T47D breast cancer cells, the predominant factor that leads to growth arrest is G1 cell cycle blockade. Our data indicate that this blockade results from the downregulation of Cyclin D1 and Cyclin D3, which in turn causes Rb hypophosphorylation. Non-toxic retinoids that are potent inducers of cell cycle arrest may be particularly useful for the prevention of breast cancer.


Journal of Biological Chemistry | 2009

Rexinoid-induced Expression of IGFBP-6 Requires RARβ-dependent Permissive Cooperation of Retinoid Receptors and AP-1

Ivan P. Uray; Qiang Shen; Hye-Sook Seo; Heetae Kim; William W. Lamph; Reid P. Bissonnette; Powel H. Brown

The synthetic rexinoid bexarotene (Targretin, LGD1069) inhibits the formation of both estrogen receptor-negative and estrogen receptor-positive breast cancer in preclinical models and controls the expression of growth-regulatory biomarkers, such as IGFBP-6 (insulin-like growth factor-binding protein 6), RARβ, or cyclin D1. In this study, we identified a classical retinoic acid-responsive element in the first intron in the IGFBP-6 gene adjacent to a consensus AP-1 binding site, both elements essential for rexinoid-induced expression of IGFBP-6. In chromatin binding experiments, bexarotene increased the occupancy of the identified enhancer element by RXRα, RARβ, cJun, cFos, and p300. In normal mammary epithelial cells and T47D breast cancer cells, small interfering RNA-mediated knockdown of all RXR isoforms or RARβ, but not RARα or RARγ alone, blocked the induction of IGFBP-6 by bexarotene. Simultaneous knockdown of RARα and RARγ abrogated both the induction of RARβ and the up-regulation and secretion of IGFBP-6. The suppression of either RARβ or cJun by small interfering RNA blocked the recruitment of RXRα and cJun to the enhancer. These results demonstrate a novel cooperative interaction between retinoid receptors and AP-1 orchestrated by RARβ and highlight a novel mechanism by which RARβ can mediate the cancer-preventive effects of rexinoids.

Collaboration


Dive into the Heetae Kim's collaboration.

Top Co-Authors

Avatar

Powel H. Brown

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Qiang Shen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ivan P. Uray

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

William W. Lamph

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Chunhua Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dianren Xia

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Harish Srinivas

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jamal Hill

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge