Network


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

Hotspot


Dive into the research topics where Yi Wen Chang is active.

Publication


Featured researches published by Yi Wen Chang.


Journal of Clinical Investigation | 2011

miR-107 promotes tumor progression by targeting the let-7 microRNA in mice and humans

Pai Sheng Chen; Jen Liang Su; Shih Ting Cha; Woan-Yuh Tarn; Ming Yang Wang; Hsing Chih Hsu; Ming-Tsan Lin; Chia-Yu Chu; Kuo-Tai Hua; Chiung-Nien Chen; Tsang Chih Kuo; King-Jen Chang; Michael Hsiao; Yi Wen Chang; Jin-Shing Chen; Pan-Chyr Yang; Min-Liang Kuo

MicroRNAs (miRNAs) influence many biological processes, including cancer. They do so by posttranscriptionally repressing target mRNAs to which they have sequence complementarity. Although it has been postulated that miRNAs can regulate other miRNAs, this has never been shown experimentally to our knowledge. Here, we demonstrate that miR-107 negatively regulates the tumor suppressor miRNA let-7 via a direct interaction. miR-107 was found to be highly expressed in malignant tissue from patients with advanced breast cancer, and its expression was inversely correlated with let-7 expression in tumors and in cancer cell lines. Ectopic expression of miR-107 in human cancer cell lines led to destabilization of mature let-7, increased expression of let-7 targets, and increased malignant phenotypes. In contrast, depletion of endogenous miR-107 dramatically increased the stability of mature let-7 and led to downregulation of let-7 targets. Accordingly, miR-107 expression increased the tumorigenic and metastatic potential of a human breast cancer cell line in mice via inhibition of let-7 and upregulation of let-7 targets. By mutating individual sites within miR-107 and let-7, we found that miR-107 directly interacts with let-7 and that the internal loop of the let-7/miR-107 duplex is critical for repression of let-7 expression. Altogether, we have identified an oncogenic role for miR-107 and provide evidence of a transregulational interaction among miRNAs in human cancer development.


Journal of Biological Chemistry | 2010

CYR61 Regulates BMP-2-dependent Osteoblast Differentiation through the αvβ3 Integrin/Integrin-linked Kinase/ERK Pathway

Jen Liang Su; Jean Chiou; Chih-Hsin Tang; Ming Zhao; Chun-Hao Tsai; Pai Sheng Chen; Yi Wen Chang; Ming Hsien Chien; Chu Ying Peng; Michael Hsiao; Ming Liang Kuo; Men Luh Yenk

Osteoporosis is one of the most common bone pathologies. A number of novel molecules have been reported to increase bone formation including cysteine-rich protein 61 (CYR61), a ligand of integrin receptor, but mechanisms remain unclear. It is known that bone morphogenetic proteins (BMPs), especially BMP-2, are crucial regulators of osteogenesis. However, the interaction between CYR61 and BMP-2 is unclear. We found that CYR61 significantly increases proliferation and osteoblastic differentiation in MC3T3-E1 osteoblasts and primary cultured osteoblasts. CYR61 enhances mRNA and protein expression of BMP-2 in a time- and dose-dependent manner. Moreover, CYR61-mediated proliferation and osteoblastic differentiation are significantly decreased by knockdown of BMP-2 expression or inhibition of BMP-2 activity. In this study we found integrin αvβ3 is critical for CYR61-mediated BMP-2 expression and osteoblastic differentiation. We also found that integrin-linked kinase, which is downstream of the αvβ3 receptor, is involved in CYR61-induced BMP-2 expression and subsequent osteoblastic differentiation through an ERK-dependent pathway. Taken together, our results show that CYR61 up-regulates BMP-2 mRNA and protein expression, resulting in enhanced cell proliferation and osteoblastic differentiation through activation of the αvβ3 integrin/integrin-linked kinase/ERK signaling pathway.


Cancer Research | 2010

Downregulation of microRNA miR-520h by E1A contributes to anticancer activity.

Jen Liang Su; Poshen B. Chen; Ya Huey Chen; Shang Chih Chen; Yi Wen Chang; Yi Hua Jan; Xiaoyun Cheng; Michael Hsiao; Mien Chie Hung

The leading cause of death in cancer patients is cancer metastasis, for which there is no effective treatment. MicroRNAs (miRNA) have been shown to play a significant role in cancer metastasis through regulation of gene expression. The adenovirus type 5 E1A (E1A) is associated with multiple tumor-suppressing activities including the inhibition of metastasis, and E1A gene therapies have been tested in several clinical trials. However, the mechanisms involved in E1A-mediated tumor-suppressing activities are not yet completely defined. Here, we showed that E1A downregulated the expression of the miRNA miR-520h, which was critical for E1A-mediated cancer cell mobility and in vitro invasion activity. In addition, we identified a signal cascade, namely, E1A-->miRNA-520h-->PP2A/C-->IkappaB kinase-->NF-kappaB-->Twist, in which E1A inhibited the expression of Twist through downregulation of miR-520h and the signal cascade. Our results indicated a functional link between miR-520h and tumorigenicity/invasive ability and provided a new insight into the role of E1A-mediated miRNA regulation in tumor suppression. Therefore, the results identified a new cascade of E1A-mediated tumor suppression activity via downregulation of miRNA-520h expression.


Journal of Clinical Investigation | 2013

Angiopoietin-like protein 1 suppresses SLUG to inhibit cancer cell motility.

Tsang Chih Kuo; Ching-Ting Tan; Yi Wen Chang; Chih Chen Hong; Wei Jiunn Lee; Min Wei Chen; Yung-Ming Jeng; Jean Chiou; Pei Yu; Pai Sheng Chen; Ming Yang Wang; Michael Hsiao; Jen Liang Su; Min-Liang Kuo

Angiopoietin-like protein 1 (ANGPTL1) is a potent regulator of angiogenesis. Growing evidence suggests that ANGPTL family proteins not only target endothelial cells but also affect tumor cell behavior. In a screen of 102 patients with lung cancer, we found that ANGPTL1 expression was inversely correlated with invasion, lymph node metastasis, and poor clinical outcomes. ANGPTL1 suppressed the migratory, invasive, and metastatic capabilities of lung and breast cancer cell lines in vitro and reduced metastasis in mice injected with cancer cell lines overexpressing ANGPTL1. Ectopic expression of ANGPTL1 suppressed the epithelial-to-mesenchymal transition (EMT) by reducing the expression of the zinc-finger protein SLUG. A microRNA screen revealed that ANGPTL1 suppressed SLUG by inducing expression of miR-630 in an integrin α(1)β(1)/FAK/ERK/SP1 pathway-dependent manner. These results demonstrate that ANGPTL1 represses lung cancer cell motility by abrogating the expression of the EMT mediator SLUG.


Cancer Research | 2011

FOXO3a-Dependent Mechanism of E1A-Induced Chemosensitization

Jen Liang Su; Xiaoyun Cheng; Hirohito Yamaguchi; Yi Wen Chang; Chao Feng Hou; Dung Fang Lee; How Wen Ko; Kuo-Tai Hua; Ying Nai Wang; Michael Hsiao; Po Shen B. Chen; Jung Mao Hsu; Robert C. Bast; Gabriel N. Hortobagyi; Mien Chie Hung

Gene therapy trials in human breast, ovarian, and head and neck tumors indicate that adenovirus E1A can sensitize cancer cells to the cytotoxic effects of paclitaxel in vitro and in vivo. Resistance to paclitaxel has been reported to occur in cells expressing low levels of the Forkhead transcription factor FOXO3a. In this article, we report that FOXO3a is critical for E1A-mediated chemosensitization to paclitaxel. RNA interference-mediated knockdown of FOXO3a abolished E1A-induced sensitivity to paclitaxel. Mechanistic investigations indicated that E1A indirectly stabilized FOXO3a by acting at an intermediate step to inhibit a ubiquitin-dependent proteolysis pathway involving the E3 ligase βTrCP and the FOXO3a inhibitory kinase IKKβ. E1A derepressed this inhibitory pathway by stimulating expression of the protein phosphatase 2A (PP2A)/C protein phosphatases, which by binding to the TGF-β-activated kinase TAK1, inhibited its ability to activate IKKβ and, thereby, to suppress βTrCP-mediated degradation of FOXO3a. Thus, by stimulating PP2A/C expression, E1A triggers a signaling cascade that stabilizes FOXO3a and mediates chemosensitization. Our findings provide a leap forward in understanding paclitaxel chemosensitization by E1A, and offer a mechanistic rational to apply E1A gene therapy as an adjuvant for improving therapeutic outcomes in patients receiving paclitaxel treatment.


Oncogene | 2016

miR-520h is crucial for DAPK2 regulation and breast cancer progression

Chih-Ming Su; Ming Yang Wang; Charlie Hong; Hsin-An Chen; Yen-Hao Su; Chih-Hsiung Wu; Ming-Te Huang; Yi Wen Chang; Shih Sheng Jiang; Shian-Ying Sung; Jang Yang Chang; Li-Tzong Chen; Pai Sheng Chen; Jen Liang Su

MicroRNAs (miRNAs) are small RNAs that suppress gene expression by their interaction with 3’untranslated region of specific target mRNAs. Although the dysregulation of miRNAs has been identified in human cancer, only a few of these miRNAs have been functionally documented in breast cancer. Thus, defining the important miRNA and functional target involved in chemoresistance is an urgent need for human breast cancer treatment. In this study, we, for the first time, identified a key role of miRNA 520h (miR-520h) in drug resistance. Through protecting cells from paclitaxel-induced apoptosis, expression of miR-520h promoted the drug resistance of human breast cancer cells. Bioinformatics prediction, compensatory mutation and functional validation further confirmed the essential role of miR-520h-suppressed Death-associated protein kinase 2 (DAPK2) expression, as restoring DAPK2 abolished miR-520h-promoted drug resistance, and knockdown of DAPK2 mitigated cell death caused by the depletion of miR-520h. Furthermore, we observed that higher level of miR-520h is associated with poor prognosis and lymph node metastasis in human breast cancer patients. These results show that miR-520h is not only an independent prognostic factor, but is also a potential functional target for future applications in cancer therapeutics.


Oncogene | 2016

Deacetylation of HSPA5 by HDAC6 leads to GP78-mediated HSPA5 ubiquitination at K447 and suppresses metastasis of breast cancer

Yi Wen Chang; C. Tseng; Ming Yang Wang; Wen Chang Chang; C-H Lee; Li-Tzong Chen; Mien Chie Hung; Jen Liang Su

Heat-shock protein 5 (HSPA5) is a marker for poor prognosis in breast cancer patients and has an important role in cancer progression, including promoting drug resistance and metastasis. In this study, we identify that the specific lysine residue 447 (K447) of HSPA5 could be modified with polyubiquitin for subsequent degradation through the ubiquitin proteasomal system, leading to the suppression of cell migration and invasion of breast cancer. We further found that GP78, an E3 ubiquitin ligase, interacted with the C-terminal region of HSPA5 and mediated HSPA5 ubiquitination and degradation. Knock down of GP78 significantly increased the expression of HSPA5 and enhanced migration/invasive ability of breast cancer cells. Knock down of histone deacetylase-6 (HDAC6) increased the acetylation of HSPA5 at lysine residues 353 (K353) and reduced GP78-mediated ubiquitination of HSPA5 at K447 and then increased cell migration/invasion. In addition, we demonstrate that E3 ubiquitin ligase GP78 preferentially binds to deacetylated HSPA5. Notably, the expression levels of GP78 inversely correlated with HSPA5 levels in breast cancer patients. Patients with low GP78 expression significantly correlated with invasiveness of breast cancer, advanced tumor stages and poor clinical outcome. Taken together, our results provide new mechanistic insights into the understanding that deacetylation of HSPA5 by HDAC6 facilitates GP78-mediated HSPA5 ubiquitination and suggest that post-translational regulation of HSPA5 protein is critical for HSPA5-mediated metastatic properties of breast cancer.


Cancer Research | 2014

MiR326 maturation is crucial for VEGF-C-driven cortactin expression and esophageal cancer progression

Chih Chen Hong; Pai Sheng Chen; Jean Chiou; Ching Feng Chiu; Ching-Yao Yang; Michael Hsiao; Yi Wen Chang; Yang Hao Yu; Mien Chie Hung; Nai Wen Hsu; Shine Gwo Shiah; Nan Yung Hsu; Jen Liang Su

Esophageal cancer is an aggressive human malignancy with increasing incidence in the developed world. VEGF-C makes crucial contributions to esophageal cancer progression that are not well understood. Here, we report the discovery of regulatory relationship in esophageal cancers between the expression of VEGF-C and cortactin (CTTN), a regulator of the cortical actin cytoskeleton. Upregulation of CTTN expression by VEGF-C enhanced the invasive properties of esophageal squamous cell carcinoma in vitro and tumor metastasis in vivo. Mechanistic investigations showed that VEGF-C increased CTTN expression by downregulating Dicer-mediated maturation of miR326, thereby relieving the suppressive effect of miR326 on CTTN expression. Clinically, expression of Dicer and miR326 correlated with poor prognosis in patients with esophageal cancer. Our findings offer insights into how VEGF-C enhances the robust invasive and metastatic properties of esophageal cancer, which has potential implications for the development of new biomarkers or therapies in this setting.


American Journal of Respiratory and Critical Care Medicine | 2015

CARMA3 Represses Metastasis Suppressor NME2 to Promote Lung Cancer Stemness and Metastasis

Yi Wen Chang; Ching Feng Chiu; Kang Yun Lee; Chih Chen Hong; Yi Yun Wang; Ching Chia Cheng; Yi Hua Jan; Ming Shyan Huang; Michael Hsiao; Jui Ti Ma; Jen Liang Su

RATIONALE CARD-recruited membrane-associated protein 3 (CARMA3) is a novel scaffold protein that regulates nuclear factor (NF)-κB activation; however, the underlying mechanism of CARMA3 in lung cancer stemness and metastasis remains largely unknown. OBJECTIVES To investigate the molecular mechanisms underlying the involvement of CARMA3 in non-small cell lung cancer progression. METHODS The expression levels of CARMA3 and NME2 in a cohort of patients with lung cancer (n = 91) were examined by immunohistochemistry staining and assessed by Kaplan-Meier survival analysis. The effects of CARMA3, microRNA-182 (miR-182), and NME2 on cancer stemness and metastasis were measured in vitro and in vivo. Chromatin immunoprecipitation and luciferase reporter assays were performed to determine the mechanisms of NF-κB-driven miR-182 expression and NME2 regulation. MEASUREMENTS AND MAIN RESULTS We observed that CARMA3 inversely correlated with NME2 expression in patients with lung cancer (Pearson correlation coefficient: R = -0.24; P = 0.022). NME2 levels were significantly decreased in tumor tissues compared with adjacent normal lung tissues (P < 0.001), and patients with lung cancer with higher levels of NME2 had longer survival outcomes (overall survival, P < 0.01; disease-free survival, P < 0.01). Mechanistically, CARMA3 promoted cell motility by reducing the level of NME2 through the NF-κB/miR-182 pathway and by increasing cancer stem cell properties and metastasis in lung cancer. CONCLUSIONS We identified a novel mechanism of CARMA3 in lung cancer stemness and metastasis through the negative regulation of NME2 by NF-κB-dependent induction of miR-182. Our findings provide an attractive strategy for targeting the CARMA3/NF-κB/miR-182 pathway as a potential treatment for lung cancer.


Annals of Surgical Oncology | 2014

Suppression of dicer increases sensitivity to gefitinib in human lung cancer cells

Jui Chieh Chen; Yen Hao Su; Ching Feng Chiu; Yi Wen Chang; Yang Hao Yu; Chi Feng Tseng; Hsin An Chen; Jen Liang Su

BackgroundAccumulating evidence is revealing an important role of microRNA (miRNA) in tumor progression and chemotherapeutic resistance. Dicer is a cytoplasmic endoribonuclease type III crucial for production of mature miRNAs. The aberrant expression of Dicer has also been reportedly associated with clinical aggressiveness, prognosis, and patient survival in various cancer types. However, the molecular mechanisms of Dicer in acquired gefitinib resistance are still not clear.MethodsIn this study, we analyzed the protein level of Dicer between gefitinib-sensitive (PC9) and gefitinib-resistant (PC9/GR) non-small-cell lung cancer (NSCLC) cell lines by Western blot analysis. Silence and overexpression of the Dicer were performed to investigate the effects on gefitinib sensitivity, as assessed by (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay and sub-G1 assay of flow cytometry. To further explore the mechanism of chemoresistance, we examined whether Dicer knockdown led to modulating specific miRNAs and its miRNA target genes.ResultsDicer expression was significantly increased in PC9/GR compared with PC9 cells. Knockdown of Dicer restores gefitinib sensitivity in resistant cells, and overexpression of Dicer enhances resistance to gefitinib in sensitive cells. Silencing of Dicer induces sensitivity to gefitinib in NSCLC cells through the downregulation of miR-30b/c and miR-221/222 to increase the protein level of caspase-3, resulting in an increase in gefitinib-induced apoptosis.ConclusionsDicer contributes to the resistance to gefitinib in lung cancer. These results indicate that Dicer may be a target for diagnosis and therapy of patients with resistance to gefitinib.

Collaboration


Dive into the Yi Wen Chang's collaboration.

Top Co-Authors

Avatar

Jen Liang Su

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mien Chie Hung

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Chih Chen Hong

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Ching Feng Chiu

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Hsin An Chen

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

Pai Sheng Chen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chih Hsiung Wu

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

Ming Yang Wang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chi Feng Tseng

National Tsing Hua University

View shared research outputs
Researchain Logo
Decentralizing Knowledge