Network


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

Hotspot


Dive into the research topics where King-Jen Chang is active.

Publication


Featured researches published by King-Jen Chang.


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.


PLOS ONE | 2009

Multiple Lineages of Human Breast Cancer Stem/Progenitor Cells Identified by Profiling with Stem Cell Markers

Wendy W. Hwang-Verslues; Wen-Hung Kuo; Po-Hao Chang; Chi-Chun Pan; Hsing-Hui Wang; Sheng Ta Tsai; Yung-Ming Jeng; Jin-Yu Shew; John T. Kung; Chung-Hsuan Chen; Eva Y.-H. P. Lee; King-Jen Chang; Wen-Hwa Lee

Heterogeneity of cancer stem/progenitor cells that give rise to different forms of cancer has been well demonstrated for leukemia. However, this fundamental concept has yet to be established for solid tumors including breast cancer. In this communication, we analyzed solid tumor cancer stem cell markers in human breast cancer cell lines and primary specimens using flow cytometry. The stem/progenitor cell properties of different marker expressing-cell populations were further assessed by in vitro soft agar colony formation assay and the ability to form tumors in NOD/SCID mice. We found that the expression of stem cell markers varied greatly among breast cancer cell lines. In MDA-MB-231 cells, PROCR and ESA, instead of the widely used breast cancer stem cell markers CD44+/CD24-/low and ALDH, could be used to highly enrich cancer stem/progenitor cell populations which exhibited the ability to self renew and divide asymmetrically. Furthermore, the PROCR+/ESA+ cells expressed epithelial-mesenchymal transition markers. PROCR could also be used to enrich cells with colony forming ability from MB-361 cells. Moreover, consistent with the marker profiling using cell lines, the expression of stem cell markers differed greatly among primary tumors. There was an association between metastasis status and a high prevalence of certain markers including CD44+/CD24−/low, ESA+, CD133+, CXCR4+ and PROCR+ in primary tumor cells. Taken together, these results suggest that similar to leukemia, several stem/progenitor cell-like subpopulations can exist in breast cancer.


Oncogene | 2007

Molecular signatures of metaplastic carcinoma of the breast by large-scale transcriptional profiling: identification of genes potentially related to epithelial-mesenchymal transition.

Huang-Chun Lien; Y. H. Hsiao; Yi-Shing Lin; Yu-Tung Yao; Hsueh-Fen Juan; Wun-Hon Kuo; Mien Chie Hung; King-Jen Chang; Fon-Jou Hsieh

Metaplastic carcinoma of the breast (MCB) is a poorly understood subtype of breast cancer. It is generally characterized by the coexistence of ductal carcinomatous and transdifferentiated sarcomatous components, but the underlying molecular alterations, possibly related to epithelial–mesenchymal transition (EMT), remain elusive. We performed transcriptional profiling using half-a-genome oligonucleotide microarrays to elucidate genetic profiles of MCBs and their differences to those of ductal carcinoma of breasts (DCBs) using discarded specimens of four MCBs and 34 DCBs. Unsupervised clustering disclosed distinctive expression profiles between MCBs and DCBs. Supervised analysis identified gene signatures discriminating MCBs from DCBs and between MCB subclasses. Notably, many of the discriminator genes were associated with downregulation of epithelial phenotypes and with synthesis, remodeling and adhesion of extracellular matrix, with some of them have known or inferred roles related to EMT. Importantly, several of the discriminator genes were upregulated in a mutant Snail-transfected MCF7 cell known to exhibit features of EMT, thereby indicating a crucial role for EMT in the pathogenesis of MCBs. Finally, the identification of SPARC and vimentin as poor prognostic factors reinforced the role of EMT in cancer progression. These data advance our understanding of MCB and offer clues to the molecular alterations underlying EMT.


Fertility and Sterility | 1995

Influences of percutaneous administration of estradiol and progesterone on human breast epithelial cell cycle in vivo

King-Jen Chang; Tigris T.Y. Lee; Gustavo Linares-Cruz; Sabine Fournier; Bruno de Ligniéres

OBJECTIVE To study the effect of E2 and P on the epithelial cell cycle of normal human breast in vivo. DESIGN Double-blind, randomized study. Topical application to the breast of a gel containing either a placebo, E2, P, or a combination of E2 and P, daily, during the 10 to 13 days preceding breast surgery. PATIENTS Forty premenopausal women undergoing breast surgery for the removal of a lump. MAIN OUTCOME MEASURES. Plasma and breast tissue concentrations of E2 and P. Epithelial cell cycle evaluated in normal breast tissue areas by counting mitoses and proliferating cell nuclear antigen immunostaining quantitative analyses. RESULTS Increased E2 concentration increases the number of cycling epithelial cells. Increased P concentration significantly decreases the number of cycling epithelial cells. CONCLUSION Exposure to P for 10 to 13 days reduces E2-induced proliferation of normal breast epithelial cells in vivo.


Oncogene | 2011

miR-495 is upregulated by E12/E47 in breast cancer stem cells, and promotes oncogenesis and hypoxia resistance via downregulation of E-cadherin and REDD1

Wendy W. Hwang-Verslues; Po-Hao Chang; Pei-Chi Wei; Ching-Yao Yang; Chun-Kai Huang; W.-H. Kuo; Jin-Yuh Shew; King-Jen Chang; Eyhp Lee; Wen-Hwa Lee

MicroRNAs (miRNAs) are involved in tumorigenecity by regulating specific oncogenes and tumor suppressor genes, and their roles in breast cancer stem cells (BCSCs) are becoming apparent. Distinct from the CD44+/CD24−/low sub-population, we have isolated a novel PROCR+/ESA+ BCSC sub-population. To explore miRNA-regulatory mechanisms in this sub-population, we performed miRNA expression profiling and found miR-495 as the most highly upegulated miRNA in PROCR+/ESA+ cells. Coincidently, high upregulation of miR-495 was also found in CD44+/CD24−/low BCSCs, reflecting its potential importance in maintaining common BCSC properties. Ectopic expression of miR-495 in breast cancer cells promoted their colony formation in vitro and tumorigenesis in mice. miR-495 directly suppressed E-cadherin expression to promote cell invasion and inhibited REDD1 expression to enhance cell proliferation in hypoxia through post-transcriptional mechanism. miR-495 expression was directly modulated by transcription factor E12/E47, which itself is highly expressed in BCSCs. These findings reveal a novel regulatory pathway centered on miR-495 that contributes to BCSC properties and hypoxia resistance.


International Journal of Cancer | 2002

High-frequency microsatellite instability predicts better chemosensitivity to high-dose 5-fluorouracil plus leucovorin chemotherapy for stage IV sporadic colorectal cancer after palliative bowel resection.

Jin-Tung Liang; Kuo-Chin Huang; Hong-Shiee Lai; Po-Huang Lee; Yung-Ming Cheng; Hey-Chi Hsu; Ann-Lii Cheng; Chih-Hung Hsu; Kun-Huei Yeh; Shih-Ming Wang; Chi Tang; King-Jen Chang

The influence of MSI on treatment outcome of colorectal cancers remains unclear and deserves further investigation. We recruited 244 patients with stage IV sporadic colorectal cancers for our study, based on appropriate eligibility criteria. Patients were nonrandomly allocated to 2 treatment groups of either with or without high‐dose 5‐FU plus leucovorin chemotherapy (HDFL, 5‐FU 2,600 mg/m2 leucovorin 300 mg/m2 maximum 500 mg). Each treatment group was further divided into 2 subgroups according to high‐frequency MSI (MSI‐H) status. MSI‐H was defined as the appearance of MSI in at least 2 of the 5 examined chromosomal loci (BAT‐25, BAT‐26, D5S346, D2S123, D17S250). We compared clinicopathologic parameters, p53 overexpression and overall survival between the groups. In addition, 4 subgroups were identified as follows: MSI‐H+HDFL+, n = 35; MSI‐H−HDFL+, n = 134; MSI‐H+HDFL−, n = 17; MSI‐H−HDFL−, n = 58. There was no significant difference of background clinicopathologic data between the HDFL+ and HDFL− treatment groups (p > 0.05). Survival analyses indicated that the patients of subgroup MSI‐H+HDFL+ survived significantly longer than those of subgroup MSI‐H−HDFL+, with median survival times of 24 (95% CI 20.2–27.9) and 13 (95% CI 11.6–14.4) months, respectively (p = 0.0001, log‐rank test). In contrast, in patients without chemotherapy, the prognosis was poor irrespective of MSI status, with median survival times of 7.0 (95% CI 4.6–9.4) and 7.0 (95% CI 6.1–7.9) months in the MSI‐H+HDFL− and MSI‐H−HDFL− subgroups, respectively (p = 0.8205, log‐rank test). MSI‐H cancers responded significantly better to HDFL (p = 0.001), with a mean response rate of 65.71% (95% CI 49.98–81.44%) in subgroup MSI‐H+HDFL+ compared to 35.07% (95% CI 26.99–43.15%) in subgroup MSI‐H−HDFL+. There appeared to be no preferential metastatic site where response to HDFL can be predicted based on the MSI status of the primary tumor. Toxicity to HDFL was similarly minimal between MSI‐H+ and MSI‐H− patients (p > 0.05). Multivariate analysis of all patients further indicated that MSI‐H and chemotherapy were independent favorable prognostic parameters (p < 0.05). Thus, the better prognosis of stage IV sporadic colorectal cancers with MSI‐H may be associated with better chemosensitivity, rather than lower aggressiveness in biologic behavior.


PLOS ONE | 2011

Breast Cancer Cells Induce Cancer-Associated Fibroblasts to Secrete Hepatocyte Growth Factor to Enhance Breast Tumorigenesis

Shiaw-Wei Tyan; Wen-Hung Kuo; Chun-Kai Huang; Chi-Chun Pan; Jin-Yuh Shew; King-Jen Chang; Eva Y.-H. P. Lee; Wen-Hwa Lee

It has been well documented that microenvironment consisting of stroma affects breast cancer progression. However, the mechanisms by which cancer cells and fibroblasts, the major cell type in stroma, interact with each other during tumor development remains to be elucidated. Here, we show that the human cancer-associated fibroblasts (CAFs) had higher activity in enhancing breast tumorigenecity compared to the normal tissue-associated fibroblasts (NAFs) isolated from the same patients. The expression level of hepatocyte growth factor (HGF) in these fibroblasts was positively correlated with their ability to enhance breast tumorigenesis in mice. Deprivation of HGF using a neutralizing antibody reduced CAF-mediated colony formation of human breast cancer cells, indicating that CAFs enhanced cancer cell colony formation mainly through HGF secretion. Co-culture with human breast cancer MDA-MB-468 cells in a transwell system enhanced NAFs to secret HGF as well as promote tumorigenecity. The newly gained ability of these “educated” NAFs became irreversible after continuing this process till fourth passage. These results suggested that breast cancer cells could alter the nature of its surrounding fibroblasts to secrete HGF to support its own progression through paracrine signaling.


Journal of Clinical Oncology | 2005

Gene Expression Profile Predicts Patient Survival of Gastric Cancer After Surgical Resection

Chiung-Nien Chen; Jen-Jen Lin; Jeremy J.W. Chen; Po-Huang Lee; Ching-Yao Yang; Min-Liang Kuo; King-Jen Chang; Fon-Jou Hsieh

PURPOSE This study was conducted to characterize gene expression profile of survival in patients with surgically curable gastric cancer by using an in-house membrane microarray and developing a survival prediction model. MATERIALS AND METHODS Data of cDNA microarrays were obtained from 18 pairs of cancerous and noncancerous gastric tissues. Nine patients who survived > 30 months were identified as good survival, and the other nine, who survived < 12 months, were identified as poor survival. Supervised analysis was performed to identify a gene expression profile by good and poor survival. Semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR) was used to confirm the microarray data in 10 patients with sufficient RNA. Using these 10 patients and another 10 patients selected randomly from 40 newly enrolled patients as the training group, the RT-PCR status of the confirmed genes was used for predicting good versus poor survival. Finally, the prediction model was tested in the remaining 30 newly enrolled gastric cancer patients. RESULTS A survival prediction model consisting of three genes (CD36, SLAM, PIM-1) was developed. This model could correctly predict poor or good survival in 23 (76.7%) of 30 newly enrolled patients, and yielded a specificity of 80% and a sensitivity of 73.3%. The survival rate of the patients predicted to have good survival was significantly higher than that of those predicted to have poor survival in the test group as a whole (N = 30; P = .00531) and in stage III patients (n = 16; P = .04467). CONCLUSION The semiquantitative RT-PCR gene expression profiling of three genes extracted from microarray study can accurately predict surgery-related outcome in gastric cancer patients.


Journal of Cell Science | 2007

CTGF enhances the motility of breast cancer cells via an integrin-αvβ3–ERK1/2-dependent S100A4-upregulated pathway

Pai Sheng Chen; Ming Yang Wang; Shin Ni Wu; Jen Liang Su; Chih Chen Hong; Shuang En Chuang; Min Wei Chen; Kuo-Tai Hua; Yu Ling Wu; Shih Ting Cha; Munisamy Suresh Babu; Chiung-Nien Chen; Po-Huang Lee; King-Jen Chang; Min-Liang Kuo

Connective tissue growth factor (CTGF) expression is elevated in advanced stages of breast cancer, but the regulatory role of CTGF in invasive breast cancer cell phenotypes is unclear. Presently, overexpression of CTGF in MCF-7 cells (MCF-7/CTGF cells) enhanced cellular migratory ability and spindle-like morphological alterations, as evidenced by actin polymerization and focal-adhesion-complex aggregation. Reducing the CTGF level in MDA-MB-231 (MDA231) cells by antisense CTGF cDNA (MDA231/AS cells) impaired cellular migration and promoted a change to an epithelial-like morphology. A neutralizing antibody against integrin αvβ3 significantly attenuated CTGF-mediated ERK1/2 activation and cellular migration, indicating that the integrin-αvβ3–ERK1/2 signaling pathway is crucial in mediating CTGF function. Moreover, the cDNA microarray analysis revealed CTGF-mediated regulation of the prometastatic gene S100A4. Transfection of MCF-7/CTGF cells with AS-S100A4 reversed the CTGF-induced cellular migratory ability, whereas overexpression of S100A4 in MDA231/AS cells restored their high migratory ability. Genetic and pharmacological manipulations suggested that the CTGF-mediated S100A4 upregulation was dependent on ERK1/2 activation, with expression levels of CTGF and S100A4 being closely correlated with human breast tumors. We conclude that CTGF plays a crucial role in migratory/invasive processes in human breast cancer by a mechanism involving activation of the integrin-αvβ3–ERK1/2–S100A4 pathway.


Breast Cancer Research and Treatment | 2006

Leptin-induced growth of human ZR-75-1 breast cancer cells is associated with up-regulation of cyclin D1 and c-Myc and down-regulation of tumor suppressor p53 and p21WAF1/CIP1.

Chiachen Chen; Yuan-Ching Chang; Chien-Liang Liu; King-Jen Chang; Ing-Cherng Guo

SummaryObesity has been recognized as a risk factor for breast cancer. Adipocyte-derived leptin may play as a paracrine regulator on the growth of breast cancer cells. Expression of both leptin and its OB-Rb receptor was detected in human breast cancer ZR-75-1 cells and further induced by leptin, suggesting that both expression and message mediation of leptin were autoregulated by itself. With cell counting and MTT assay, we had observed leptin stimulated ZR-75-1 growth in dose- and time-dependent manners. To study what steps of cell cycle progression leptin may involve in, we analyzed cell-cycle profile with flow cytometric analysis, mRNA and protein expressions of four cell-cycle regulators with RT-PCR and Western blotting analysis. Under the treatment of leptin, the G1 arrest of cells was reduced accompanied with up-regulation of G1 phase-specific cyclin D1 and proto-oncogene c-Myc, but down-regulation of cyclin-dependent kinase inhibitor p21WAF1/CIP1 and tumor suppressor p53. Furthermore, JAK2 inhibitor AG490, PI3K/Akt inhibitor Wortmannin, and MEK/ERK1/2 inhibitor PD98059 were efficiently prevented leptin-promoted cell growth. Effect of cooperation between leptin and estrogen on ZR-75-1 growth had been observed. Collectively, the results showed that the proliferative effect of leptin on ZR-75-1 was associated with the up-regulation of cyclin D1 and c-Myc and down-regulation of tumor suppressor p53 and p21WAF1/CIP1 plausibly through a hypothesized JAK2-PI3K/Akt-MEK/ERK pathway. The leptin- and OB-Rb-expressing capability of ZR-75-1 created a possible autocrine control of leptin, in which signal could be effectively amplified by itself, on cell growth.

Collaboration


Dive into the King-Jen Chang's collaboration.

Top Co-Authors

Avatar

Po-Huang Lee

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Wen-Hung Kuo

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chiung-Nien Chen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chiun-Sheng Huang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Jin-Tung Liang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Fon-Jou Hsieh

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Yung-Ming Jeng

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Min-Liang Kuo

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Ming-Tsan Lin

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Hong-Shiee Lai

National Taiwan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge