Network


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

Hotspot


Dive into the research topics where Mien Chie Hung is active.

Publication


Featured researches published by Mien Chie Hung.


Nature Cell Biology | 2004

Dual regulation of Snail by GSK-3β-mediated phosphorylation in control of epithelial–mesenchymal transition

Binhua P. Zhou; Jiong Deng; Weiya Xia; Jihong Xu; Yan M. Li; Mehmet Gunduz; Mien Chie Hung

The phenotypic changes of increased motility and invasiveness of cancer cells are reminiscent of the epithelial–mesenchymal transition (EMT) that occurs during embryonic development. Snail, a zinc-finger transcription factor, triggers this process by repressing E-cadherin expression; however, the mechanisms that regulate Snail remain elusive. Here we find that Snail is highly unstable, with a short half-life about 25 min. We show that GSK-3β binds to and phosphorylates Snail at two consensus motifs to dually regulate the function of this protein. Phosphorylation of the first motif regulates its β-Trcp-mediated ubiquitination, whereas phosphorylation of the second motif controls its subcellular localization. A variant of Snail (Snail-6SA), which abolishes these phosphorylations, is much more stable and resides exclusively in the nucleus to induce EMT. Furthermore, inhibition of GSK-3β results in the upregulation of Snail and downregulation of E-cadherin in vivo. Thus, Snail and GSK-3β together function as a molecular switch for many signalling pathways that lead to EMT.


Nature Cell Biology | 2001

Cytoplasmic localization of p21 Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu -overexpressing cells

Binhua P. Zhou; Yong Liao; Weiya Xia; Bill Spohn; Mong-Hong Lee; Mien Chie Hung

Amplification or overexpression of HER-2/neu in cancer cells confers resistance to apoptosis and promotes cell growth. The cellular localization of p21Cip1/WAF1 has been proposed to be critical either in promoting cell survival or in inhibiting cell growth. Here we show that HER-2/neu-mediated cell growth requires the activation of Akt, which associates with p21Cip1/WAF1 and phosphorylates it at threonine 145, resulting in cytoplasmic localization of p21Cip1/WAF1. Furthermore, blocking the Akt pathway with a dominant-negative Akt mutant restores the nuclear localization and cell-growth-inhibiting activity of p21Cip1/WAF1. Our results indicate that HER-2/neu induces cytoplasmic localization of p21Cip1/WAF1 through activation of Akt to promote cell growth, which may have implications for the oncogenic activity of HER-2/neu and Akt.


Nature Cell Biology | 2001

Nuclear localization of EGF receptor and its potential new role as a transcription factor

Shiaw Yih Lin; Keishi Makino; Weiya Xia; Angabin Matin; Yong Wen; Ka Yin Kwong; Lilly Y. W. Bourguignon; Mien Chie Hung

Epidermal growth factor receptor (EGFR) has been detected in the nucleus in many tissues and cell lines. However, the potential functions of nuclear EGFR have largely been overlooked. Here we demonstrate that nuclear EGFR is strongly correlated with highly proliferating activities of tissues. When EGFR was fused to the GAL4 DNA-binding domain, we found that the carboxy terminus of EGFR contained a strong transactivation domain. Moreover, the receptor complex bound and activated AT-rich consensus-sequence-dependent transcription, including the consensus site in cyclin D1 promoter. By using chromatin immunoprecipitation assays, we further demonstrated that nuclear EGFR associated with promoter region of cyclin D1 in vivo. EGFR might therefore function as a transcription factor to activate genes required for highly proliferating activities.


Nature Cell Biology | 2001

HER-2/neu induces p53 ubiquitination via Akt-mediated MDM2 phosphorylation.

Binhua P. Zhou; Yong Liao; Weiya Xia; Yiyu Zou; Bill Spohn; Mien Chie Hung

HER-2/neu amplification or overexpression can make cancer cells resistant to apoptosis and promotes their growth. p53 is crucial in regulating cell growth and apoptosis, and is often mutated or deleted in many types of tumour. Moreover, many tumours with a wild-type gene for p53 do not have normal p53 function, suggesting that some oncogenic signals suppress the function of p53. In this study, we show that HER-2/neu-mediated resistance to DNA-damaging agents requires the activation of Akt, which enhances MDM2-mediated ubiquitination and degradation of p53. Akt physically associates with MDM2 and phosphorylates it at Ser166 and Ser186. Phosphorylation of MDM2 enhances its nuclear localization and its interaction with p300, and inhibits its interaction with p19ARF, thus increasing p53 degradation. Our study indicates that blocking the Akt pathway mediated by HER-2/neu would increase the cytotoxic effect of DNA-damaging drugs in tumour cells with wild-type p53.


Cancer Research | 2008

An Integrative Genomic and Proteomic Analysis of PIK3CA, PTEN, and AKT Mutations in Breast Cancer

Katherine Stemke-Hale; Ana M. Gonzalez-Angulo; Ana Lluch; Richard M. Neve; Wen Lin Kuo; Michael Davies; Mark S. Carey; Zhi Hu; Yinghui Guan; Aysegul A. Sahin; W. Fraser Symmans; Lajos Pusztai; Laura K. Nolden; Hugo M. Horlings; Katrien Berns; Mien Chie Hung; Marc J. van de Vijver; Vicente Valero; Joe W. Gray; René Bernards; Gordon B. Mills; Bryan T. Hennessy

Phosphatidylinositol 3-kinase (PI3K)/AKT pathway aberrations are common in cancer. By applying mass spectroscopy-based sequencing and reverse-phase protein arrays to 547 human breast cancers and 41 cell lines, we determined the subtype specificity and signaling effects of PIK3CA, AKT, and PTEN mutations and the effects of PIK3CA mutations on responsiveness to PI3K inhibition in vitro and on outcome after adjuvant tamoxifen. PIK3CA mutations were more common in hormone receptor-positive (34.5%) and HER2-positive (22.7%) than in basal-like tumors (8.3%). AKT1 (1.4%) and PTEN (2.3%) mutations were restricted to hormone receptor-positive cancers. Unlike AKT1 mutations that were absent from cell lines, PIK3CA (39%) and PTEN (20%) mutations were more common in cell lines than tumors, suggesting a selection for these but not AKT1 mutations during adaptation to culture. PIK3CA mutations did not have a significant effect on outcome after adjuvant tamoxifen therapy in 157 hormone receptor-positive breast cancer patients. PIK3CA mutations, in comparison with PTEN loss and AKT1 mutations, were associated with significantly less and inconsistent activation of AKT and of downstream PI3K/AKT signaling in tumors and cell lines. PTEN loss and PIK3CA mutation were frequently concordant, suggesting different contributions to pathophysiology. PTEN loss rendered cells significantly more sensitive to growth inhibition by the PI3K inhibitor LY294002 than did PIK3CA mutations. Thus, PI3K pathway aberrations likely play a distinct role in the pathogenesis of different breast cancer subtypes. The specific aberration present may have implications for the selection of PI3K-targeted therapies in hormone receptor-positive breast cancer.


Journal of Clinical Oncology | 2009

Metformin and Pathologic Complete Responses to Neoadjuvant Chemotherapy in Diabetic Patients With Breast Cancer

Sao Jiralerspong; Shana L. Palla; Sharon H. Giordano; Funda Meric-Bernstam; Cornelia Liedtke; Chad M. Barnett; Limin Hsu; Mien Chie Hung; Gabriel N. Hortobagyi; Ana M. Gonzalez-Angulo

PURPOSE Population studies have suggested that metformin use in diabetic patients decreases cancer incidence and mortality. Metformin inhibits the growth of cancer cells in vitro and tumors in vivo. However, there is little clinical data to support this. Our purpose was to determine whether metformin use was associated with a change in pathologic complete response (pCR) rates in diabetic patients with breast cancer receiving neoadjuvant chemotherapy. PATIENTS AND METHODS We identified 2,529 patients who received neoadjuvant chemotherapy for early-stage breast cancer between 1990 and 2007. Patients were compared by groups: 68 diabetic patients taking metformin, 87 diabetic patients not taking metformin, and 2,374 nondiabetic patients. pCR rates were compared between the three groups using chi(2) tests of independence and compared pair- wise using a binomial test of proportions. Factors predictive of pCR were assessed using a multivariate logistic regression model. RESULTS The rate of pCR was 24% in the metformin group, 8.0% in the nonmetformin group, and 16% in the nondiabetic group (P = .02). Pairwise comparisons between the metformin and nonmetformin groups (P = .007) and the nonmetformin and nondiabetic groups (P = .04) were significant. Comparison of the pCR rates between the metformin and nondiabetic groups trended toward but did not meet significance (P = .10). Metformin use was independently predictive of pCR (odds ratio, 2.95; P = .04) after adjustment for diabetes, body mass index, age, stage, grade, receptor status, and neoadjuvant taxane use. CONCLUSION Diabetic patients with breast cancer receiving metformin and neoadjuvant chemotherapy have a higher pCR rate than do diabetics not receiving metformin. Additional studies to evaluate the potential of metformin as an antitumor agent are warranted.


Nature Cell Biology | 2011

p53 regulates epithelial–mesenchymal transition and stem cell properties through modulating miRNAs

Chun-Ju Chang; Chi Hong Chao; Weiya Xia; Jer Yen Yang; Yan Xiong; Chia Wei Li; Wen Hsuan Yu; Sumaiyah K. Rehman; Jennifer L. Hsu; Heng Huan Lee; Mo Liu; Chun Te Chen; Dihua Yu; Mien Chie Hung

The epithelial–mesenchymal transition (EMT) has recently been linked to stem cell phenotype. However, the molecular mechanism underlying EMT and regulation of stemness remains elusive. Here, using genomic approaches, we show that tumour suppressor p53 has a role in regulating both EMT and EMT-associated stem cell properties through transcriptional activation of the microRNA miR-200c. p53 transactivates miR-200c through direct binding to the miR-200c promoter. Loss of p53 in mammary epithelial cells leads to decreased expression of miR-200c and activates the EMT programme, accompanied by an increased mammary stem cell population. Re-expressing miR-200c suppresses genes that mediate EMT and stemness properties and thereby reverts the mesenchymal and stem-cell-like phenotype caused by loss of p53 to a differentiated epithelial cell phenotype. Furthermore, loss of p53 correlates with a decrease in the level of miR-200c, but an increase in the expression of EMT and stemness markers, and development of a high tumour grade in a cohort of breast tumours. This study elucidates a role for p53 in regulating EMT–MET (mesenchymal–epithelial transition) and stemness or differentiation plasticity, and reveals a potential therapeutic implication to suppress EMT-associated cancer stem cells through activation of the p53–miR-200c pathway.


Cell | 2007

IKKβ Suppression of TSC1 Links Inflammation and Tumor Angiogenesis via the mTOR Pathway

Dung Fang Lee; Hsu Ping Kuo; Chun Te Chen; Jung Mao Hsu; Chao Kai Chou; Yongkun Wei; Hui Lung Sun; Long Yuan Li; Bo Ping; Wei Chien Huang; Xianghuo He; Jen Yu Hung; Chien-Chen Lai; Qingqing Ding; Jen Liang Su; Jer Yen Yang; Aysegul A. Sahin; Gabriel N. Hortobagyi; Fuu Jen Tsai; Chang Hai Tsai; Mien Chie Hung

TNFalpha has recently emerged as a regulator linking inflammation to cancer pathogenesis, but the detailed cellular and molecular mechanisms underlying this link remain to be elucidated. The tuberous sclerosis 1 (TSC1)/TSC2 tumor suppressor complex serves as a repressor of the mTOR pathway, and disruption of TSC1/TSC2 complex function may contribute to tumorigenesis. Here we show that IKKbeta, a major downstream kinase in the TNFalpha signaling pathway, physically interacts with and phosphorylates TSC1 at Ser487 and Ser511, resulting in suppression of TSC1. The IKKbeta-mediated TSC1 suppression activates the mTOR pathway, enhances angiogenesis, and results in tumor development. We further find that expression of activated IKKbeta is associated with TSC1 Ser511 phosphorylation and VEGF production in multiple tumor types and correlates with poor clinical outcome of breast cancer patients. Our findings identify a pathway that is critical for inflammation-mediated tumor angiogenesis and may provide a target for clinical intervention in human cancer.


Cancer Research | 2007

Epidermal Growth Factor Receptor Cooperates with Signal Transducer and Activator of Transcription 3 to Induce Epithelial-Mesenchymal Transition in Cancer Cells via Up-regulation of TWIST Gene Expression

Hui-Wen Lo; Sheng-Chieh Hsu; Weiya Xia; Xinyu Cao; Jin-Yuan Shih; Yongkun Wei; James L. Abbruzzese; Gabriel N. Hortobagyi; Mien Chie Hung

Aberrant epidermal growth factor receptor (EGFR) signaling is a major cause of tumor progression and metastasis; the underlying mechanisms, however, are not well understood. In particular, it remains elusive whether deregulated EGFR pathway is involved in epithelial-mesenchymal transition (EMT), an early event that occurs during metastasis of cancers of an epithelial origin. Here, we show that EGF induces EGFR-expressing cancer cells to undergo a transition from the epithelial to the spindle-like mesenchymal morphology. EGF reduced E-cadherin expression and increased that of mesenchymal proteins. In search of a downstream mediator that may account for EGF-induced EMT, we focused on transcription repressors of E-cadherin, TWIST, SLUG, and Snail and found that cancer cells express high levels of TWIST and that EGF enhances its expression. EGF significantly increases TWIST transcripts and protein in EGFR-expressing lines. Forced expression of EGFR reactivates TWIST expression in EGFR-null cells. TWIST expression is suppressed by EGFR and Janus-activated kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) inhibitors, but not significantly by those targeting phosphoinositide-3 kinase and MEK/ERK. Furthermore, constitutively active STAT3 significantly activates the TWIST promoter, whereas the JAK/STAT3 inhibitor and dominant-negative STAT3 suppressed TWIST promoter. Deletion/mutation studies further show that a 26-bp promoter region contains putative STAT3 elements required for the EGF-responsiveness of the TWIST promoter. Chromatin immunoprecipitation assays further show that EGF induces binding of nuclear STAT3 to the TWIST promoter. Immunohistochemical analysis of 130 primary breast carcinomas indicates positive correlations between non-nuclear EGFR and TWIST and between phosphorylated STAT3 and TWIST. Together, we report here that EGF/EGFR signaling pathways induce cancer cell EMT via STAT3-mediated TWIST gene expression.


Nature Cell Biology | 2008

ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation

Jer Yen Yang; Cong S. Zong; Weiya Xia; Hirohito Yamaguchi; Qingqing Ding; Xiaoming Xie; Jing Yu Lang; Chien-Chen Lai; Chun-Ju Chang; Wei Chien Huang; Hsin Huang; Hsu Ping Kuo; Dung Fang Lee; Long Yuan Li; Huang-Chun Lien; Xiaoyun Cheng; King-Jen Chang; Chwan-Deng Hsiao; Fuu Jen Tsai; Chang Hai Tsai; Aysegul A. Sahin; William J. Muller; Gordon B. Mills; Dihua Yu; Gabriel N. Hortobagyi; Mien Chie Hung

The RAS–ERK pathway is known to play a pivotal role in differentiation, proliferation and tumour progression. Here, we show that Erk downregulates Forkhead box O 3a (FOXO3a) by directly interacting with and phosphorylating FOXO3a at Ser 294, Ser 344 and Ser 425, which consequently promotes cell proliferation and tumorigenesis. The ERK-phosphorylated FOXO3a degrades via an MDM2-mediated ubiquitin-proteasome pathway. However, the non-phosphorylated FOXO3a mutant is resistant to the interaction and degradation by murine double minute 2 (MDM2), thereby resulting in a strong inhibition of cell proliferation and tumorigenicity. Taken together, our study elucidates a novel pathway in cell growth and tumorigenesis through negative regulation of FOXO3a by RAS–ERK and MDM2.

Collaboration


Dive into the Mien Chie Hung's collaboration.

Top Co-Authors

Avatar

Weiya Xia

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dihua Yu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gabriel N. Hortobagyi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jennifer L. Hsu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yongkun Wei

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hirohito Yamaguchi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dung Fang Lee

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Chun Te Chen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Chao Kai Chou

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shao Chun Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge