Network


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

Hotspot


Dive into the research topics where Miao-Juei Huang is active.

Publication


Featured researches published by Miao-Juei Huang.


Cancer Research | 2011

Mucin glycosylating enzyme GALNT2 regulates the malignant character of hepatocellular carcinoma by modifying the EGF receptor.

Yao-Ming Wu; Chiung-Hui Liu; Rey-Heng Hu; Miao-Juei Huang; Jian-Jr Lee; Chi-Hau Chen; John Huang; Hong-Shiee Lai; Po-Huang Lee; Wen-Ming Hsu; Hsiu-Chin Huang; Min-Chuan Huang

Extracellular glycosylation is a critical determinant of malignant character. Here, we report that N-acetylgalactosaminyltransferase 2 (GALNT2), the enzyme that mediates the initial step of mucin type-O glycosylation, is a critical mediator of malignant character in hepatocellular carcinoma (HCC) that acts by modifying the activity of the epidermal growth factor receptor (EGFR). GALNT2 mRNA and protein were downregulated frequently in HCC tumors where these events were associated with vascular invasion and recurrence. Restoring GALNT2 expression in HCC cells suppressed EGF-induced cell growth, migration, and invasion in vitro and in vivo. Mechanistic investigations revealed that the status of the O-glycans attached to the EGFR was altered by GALNT2, changing EGFR responses after EGF binding. Inhibiting EGFR activity with erlotinib decreased the malignant characters caused by siRNA-mediated knockdown of GALNT2 in HCC cells, establishing the critical role of EGFR in mediating the effects of GALNT2 expression. Taken together, our results suggest that GALNT2 dysregulation contributes to the malignant behavior of HCC cells, and they provide novel insights into the significance of O-glycosylation in EGFR activity and HCC pathogenesis.


Cancer Research | 2013

C1GALT1 Enhances Proliferation of Hepatocellular Carcinoma Cells via Modulating MET Glycosylation and Dimerization

Yao-Ming Wu; Chiung-Hui Liu; Miao-Juei Huang; Hong-Shiee Lai; Po-Huang Lee; Rey-Heng Hu; Min-Chuan Huang

Altered glycosylation is a hallmark of cancer. The core 1 β1,3-galactosyltransferase (C1GALT1) controls the formation of mucin-type O-glycans, far overlooked and underestimated in cancer. Here, we report that C1GALT1 mRNA and protein are frequently overexpressed in hepatocellular carcinoma tumors compared with nontumor liver tissues, where it correlates with advanced tumor stage, metastasis, and poor survival. Enforced expression of C1GALT1 was sufficient to enhance cell proliferation, whereas RNA interference-mediated silencing of C1GALT1 was sufficient to suppress cell proliferation in vitro and in vivo. Notably, C1GALT1 attenuation also suppressed hepatocyte growth factor (HGF)-mediated phosphorylation of the MET kinase in hepatocellular carcinoma cells, whereas enforced expression of C1GALT1 enhanced MET phosphorylation. MET blockade with PHA665752 inhibited C1GALT1-enhanced cell viability. In support of these results, we found that the expression level of phospho-MET and C1GALT1 were associated in primary hepatocellular carcinoma tissues. Mechanistic investigations showed that MET was decorated with O-glycans, as revealed by binding to Vicia villosa agglutinin and peanut agglutinin. Moreover, C1GALT1 modified the O-glycosylation of MET, enhancing its HGF-induced dimerization and activation. Together, our results indicate that C1GALT1 overexpression in hepatocellular carcinoma activates HGF signaling via modulation of MET O-glycosylation and dimerization, providing new insights into how O-glycosylation drives hepatocellular carcinoma pathogenesis.


Oral Oncology | 2014

GALNT2 enhances migration and invasion of oral squamous cell carcinoma by regulating EGFR glycosylation and activity

Mei-Chun Lin; Miao-Juei Huang; Chiung-Hui Liu; Tsung-Lin Yang; Min-Chuan Huang

OBJECTIVES Oral squamous cell carcinoma (OSCC) is one of the leading cancers worldwide. Aberrant glycosylation affects many cellular properties in cancers, including OSCC. This study aimed to explore the role of N-acetylgalactosaminyltransferase 2 (GALNT2) in OSCC. MATERIALS AND METHODS Immunohistochemistry was performed to study the expression of GALNT2 in an OSCC tissue microarray. Effects of GALNT2 overexpression and knockdown on cell migration and invasion were analyzed in SAS cells by transwell migration assay and matrigel invasion assay, respectively. The Vicia villosa agglutinin (VVA) pull down assay was conducted to detect changes in O-glycans on acceptor substrates of GALNT2. Cell signaling was analyzed by Western blotting. RESULTS GALNT2 was overexpressed in 73% (35/48) of OSCC tissues. Moreover, GALNT2 expression was localized in the invasive front and increased in high grade OSCC. GALNT2 overexpression enhanced migration and invasion of SAS cells triggered by fetal bovine serum (FBS) and epidermal growth factor (EGF). In contrast, GALNT2 knockdown inhibited SAS cell migration and invasion. Furthermore, GALNT2 overexpression enhanced VVA binding to epidermal growth factor receptor (EGFR) and EGF-induced phosphorylation of EGFR and AKT. Conversely, GALNT2 knockdown decreased VVA binding and suppressed activity of EGFR and AKT. CONCLUSION GALNT2 is frequently overexpressed in OSCC, especially in the carcinoma cells at the invasive front. GALNT2 overexpression enhances the invasive potential of OSCC cells via modifying O-glycosylation and activity of EGFR. These findings suggest that GALNT2 plays an important role in the invasive behavior of OSCC and that targeting GALNT2 could be a promising approach for OSCC therapy.


Journal of Applied Physics | 1999

Optical properties of Si-doped GaN films

H. C. Yang; Tai-Yuan Lin; Miao-Juei Huang; Yang-Fang Chen

Results of the front-side and back-side photoluminescence (PL) measurements in a set of Si-doped GaN epifilms are presented. From the back-side PL spectrum, the enhancement of the yellow emission implies that most of the intrinsic defects responsible for the yellow band exist mainly near the interface between the buffer layer and the epilayer. We also found that the intensity of the yellow luminescence decreases with increasing Si dopants, which is consistent with the fact that the microscopic origin of the yellow emission can be attributed to gallium vacancies VGa. In additions, our investigations reveal that the potential fluctuations, that give rise to the effect of band-gap narrowing and linewidth broadening, are mainly caused by randomly distributed doping impurities instead of other defects.


The Journal of Clinical Endocrinology and Metabolism | 2011

MUC1 Expression Is Elevated in Severe Preeclamptic Placentas and Suppresses Trophoblast Cell Invasion via β1-Integrin Signaling

Ming-Kwang Shyu; Chih-Wei Chen; Neng-Yu Lin; Wen-Chieh Liao; Chi-Hau Chen; Chun-Jung Lin; Hsiu-Chin Huang; Jian-Jr Lee; Miao-Juei Huang; Guo-Fang Tseng; Jin-Chung Shih; Chien-Nan Lee; Fon-Jou Hsieh; Min-Chuan Huang

CONTEXT Preeclampsia is a pregnancy-specific disorder that features insufficient extravillous trophoblast (EVT) invasion. We have previously shown that MUC1 expression in human placenta increases with gestational age and inhibits choriocarcinoma cell invasion. OBJECTIVE Here, we studied whether MUC1 expression in preeclamptic placentas is dysregulated and the mechanism of EVT invasion regulated by MUC1. DESIGN MUC1 expression in severe preeclamptic placentas and gestational age-matched control placentas was analyzed by real-time RT-PCR, Western blot analysis, and immunohistochemistry. The effects of MUC1 expression on cell-matrix adhesion, invasion, and cell signaling were studied in HTR8/SVneo EVT cells. RESULTS We found that MUC1 mRNA and MUC1 protein were significantly up-regulated in severe preeclamptic placentas when compared with the gestational age-matched control placentas. Immunohistochemical analyses showed increased expression of MUC1 in the syncytiotrophoblast and EVT of severe preeclamptic placentas. In addition, MUC1 overexpression suppressed cell-matrix adhesion and invasion of EVT cells. Importantly, our data showed that MUC1 overexpression inhibited β1-integrin activity and phosphorylation of focal adhesion kinase, whereas the surface expression of β1-integrin was not significantly changed. CONCLUSIONS Our findings suggest that MUC1 is overexpressed in severe preeclamptic placentas and that MUC1 overexpression suppresses EVT invasion mainly via modulating β1-integrin signaling.


Clinical Cancer Research | 2013

β-1,4-Galactosyltransferase III enhances invasive phenotypes via β1-integrin and predicts poor prognosis in neuroblastoma.

Hsiu-Hao Chang; Chia-Hua Chen; Chih-Hsing Chou; Yung-Feng Liao; Miao-Juei Huang; Ya-Hsin Chen; Wei-Jen Wang; John Huang; Ji-Shiang Hung; Wan-Ling Ho; Yung-Ming Jeng; Mei-Ieng Che; Hsinyu Lee; Meng-Yao Lu; Yung-Li Yang; Shiann-Tarng Jou; Dong-Tsamn Lin; Kai-Hsin Lin; Wen-Ming Hsu; Min-Chuan Huang

Purpose: Neuroblastoma (NB) is a neural crest-derived tumor that commonly occurs in childhood. β-1,4-Galactosyltransferase III (B4GALT3) is highly expressed in human fetal brain and is responsible for the generation of poly-N-acetyllactosamine, which plays a critical role in tumor progression. We therefore investigated the expression and role of B4GALT3 in NB. Experimental Design: We examined B4GALT3 expression in tumor specimens from 101 NB patients by immunohistochemistry and analyzed the correlation between B4GALT3 expression and clinicopathologic factors or survival. The functional role of B4GALT3 expression was investigated by overexpression or knockdown of B4GALT3 in NB cells for in vitro and in vivo studies. Results: We found that B4GALT3 expression correlated with advanced clinical stages (P = 0.040), unfavorable Shimada histology (P < 0.001), and lower survival rate (P < 0.001). Multivariate analysis showed that B4GALT3 expression is an independent prognostic factor for poor survival of NB patients. B4GALT3 overexpression increased migration, invasion, and tumor growth of NB cells, whereas B4GALT3 knockdown suppressed the malignant phenotypes of NB cells. Mechanistic investigation showed that B4GALT3-enhanced migration and invasion were significantly suppressed by β1-integrin blocking antibody. Furthermore, B4GALT3 overexpression increased lactosamine glycans on β1-integrin, increased expression of mature β1-integrin via delayed degradation, and enhanced phosphorylation of focal adhesion kinase. Conversely, these properties were decreased by knockdown of B4GALT3 in NB cells. Conclusions: Our findings suggest that B4GALT3 predicts an unfavorable prognosis for NB and may regulate invasive phenotypes through modulating glycosylation, degradation, and signaling of β1-integrin in NB cells. Clin Cancer Res; 19(7); 1705–16. ©2013 AACR.


PLOS ONE | 2013

COSMC Is Overexpressed in Proliferating Infantile Hemangioma and Enhances Endothelial Cell Growth via VEGFR2

Jian-Jr Lee; Chia-Hua Chen; Ya-Hsin Chen; Miao-Juei Huang; John Huang; Ji-Shiang Hung; Ming-Ting Chen; Min-Chuan Huang

Infantile hemangiomas are localized lesions comprised primarily of aberrant endothelial cells. COSMC plays a crucial role in blood vessel formation and is characterized as a molecular chaperone of T-synthase which catalyzes the synthesis of T antigen (Galβ1,3GalNAc). T antigen expression is associated with tumor malignancy in many cancers. However, roles of COSMC in infantile hemangioma are still unclear. In this study, immunohistochemistry showed that COSMC was upregulated in proliferating hemangiomas compared with involuted hemangiomas. Higher levels of T antigen expression were also observed in the proliferating hemangioma. Overexpression of COSMC significantly enhanced cell growth and phosphorylation of AKT and ERK in human umbilical vein endothelial cells (HUVECs). Conversely, knockdown of COSMC with siRNA inhibited endothelial cell growth. Mechanistic investigation showed that O-glycans were present on VEGFR2 and these structures were modulated by COSMC. Furthermore, VEGFR2 degradation was delayed by COSMC overexpression and facilitated by COSMC knockdown. We also showed that COSMC was able to regulate VEGF-triggered phosphorylation of VEGFR2. Our results suggest that COSMC is a novel regulator for VEGFR2 signaling in endothelial cells and dysregulation of COSMC expression may contribute to the pathogenesis of hemangioma.


PLOS ONE | 2014

C1GALT1 Promotes Invasive Phenotypes of Hepatocellular Carcinoma Cells by Modulating Integrin β1 Glycosylation and Activity

Chiung-Hui Liu; Rey-Heng Hu; Miao-Juei Huang; I-Rue Lai; Chia-Hua Chen; Hong-Shiee Lai; Yao-Ming Wu; Min-Chuan Huang

Cancer cell invasion and metastasis are the primary causes of treatment failure and death in hepatocellular carcinoma (HCC). We previously reported that core 1 β1,3-galactosyltransferase (C1GALT1) is frequently overexpressed in HCC tumors and its expression is associated with advanced tumor stage, metastasis, and poor survival. However, the underlying mechanisms of C1GALT1 in HCC malignancy remain unclear. In this study, we found that overexpression of C1GALT1 enhanced HCC cell adhesion to extracellular matrix (ECM) proteins, migration, and invasion, whereas RNAi-mediated knockdown of C1GALT1 suppressed these phenotypes. The promoting effect of C1GALT1 on the metastasis of HCC cells was demonstrated in a mouse xenograft model. Mechanistic investigations showed that the C1GALT1-enhanced phenotypic changes in HCC cells were significantly suppressed by anti-integrin β1 blocking antibody. Moreover, C1GALT1 was able to modify O-glycans on integrin β1 and regulate integrin β1 activity as well as its downstream signaling. These results suggest that C1GALT1 could enhance HCC invasiveness through integrin β1 and provide novel insights into the roles of O-glycosylation in HCC metastasis.


Gynecologic Oncology | 2016

MUC20 promotes aggressive phenotypes of epithelial ovarian cancer cells via activation of the integrin β1 pathway

Chi-Hau Chen; Ming-Kwang Shyu; Wang Sm; Chih-Hsing Chou; Miao-Juei Huang; Tzu-Chi Lin; Syue-Ting Chen; Ho-Hsiung Lin; Min-Chuan Huang

OBJECTIVE Mucin (MUC) 20 has recently been implicated to play a role in human carcinogenesis. However, the role of MUC20 in epithelial ovarian cancer (EOC) remains to be elucidated. METHODS MUC20 expression was assessed in tissue microarray and tumor specimens of EOC patients by immunohistochemistry. Effects of MUC20 on cell viability, adhesion, migration, and invasion were analyzed in MUC20 overexpressing or knockdown EOC cells. Western blotting was performed to analyze signaling pathways modulated by MUC20. RESULTS MUC20 was overexpressed in EOC samples compared with benign tissues. High MUC20 expression was significantly associated with poor overall survival in patients with advanced-stage disease. MUC20 overexpression significantly enhanced EOC cell migration and invasion, but not viability. Mechanistic investigations showed that MUC20 increased cell adhesion to extracellular matrix (ECM) proteins and enhanced activation of integrin β1 and phosphorylation of focal adhesion kinase (FAK). The enhancement of cell motility and the integrin β1 signaling by MUC20 was significantly suppressed by integrin β1 blocking antibody. Furthermore, these effects of MUC20 on EOC cells were also demonstrated in MUC20 knockdown cells. CONCLUSIONS Our results suggest that MUC20 enhances aggressive behaviors of EOC cells by activating integrin β1 signaling and provide novel insights into the role of MUC20 in ovarian cancer metastasis.


International Journal of Gynecological Cancer | 2017

C1GALT1 Seems to Promote In Vitro Disease Progression in Ovarian Cancer

Chih-Hsing Chou; Miao-Juei Huang; Ying-Yu Liao; Chi-Hau Chen; Min-Chuan Huang

Objective Aberrant glycosylation affects many cellular properties in cancers. The core 1 &bgr;1,3-galactosyltransferase (C1GALT1), an enzyme that controls the formation of mucin-type O-glycans, has been reported to regulate hepatocellular and mammary carcinogenesis. This study aimed to explore the role of C1GALT1 in ovarian cancer. Methods C1GALT1 expression was assessed in a public database based on microarray data from 1287 ovarian cancer patients and ovarian cancerous tissues. Lectin blotting and flow cytometry analysis were conducted to detect changes in O-glycans on ovarian cancer cells. Effects of C1GALT1 on cell growth, migration, and sphere formation were analyzed in C1GALT1 knockdown or overexpressing ovarian cancer cells in vitro. Expression of cancer stemness-related genes was analyzed by quantitative reverse transcription polymerase chain reaction. Results High C1GALT1 expression shows a trend toward association with poor survival in ovarian cancer patients. C1GALT1 modifies O-glycan expression on surfaces and glycoproteins of ovarian cancer cells. Knockdown of C1GALT1 decreased cell growth, migration, and sphere formation of ES-2 and OVTW59-p4 cells. Conversely, overexpression of C1GALT1 promoted such malignant properties of SKOV3 cells. Furthermore, C1GALT1 regulated the expression of several cancer stemness-related genes, including CD133, CD24, Oct4, Nanog, and SNAI2, in ovarian cancer cells. Conclusions C1GALT1 modifies O-glycan expression and enhances malignant behaviors in ovarian cancer cells, suggesting that C1GALT1 plays a role in the pathogenesis of ovarian cancer and targeting C1GALT1 could be a promising approach for ovarian cancer therapy.

Collaboration


Dive into the Miao-Juei Huang's collaboration.

Top Co-Authors

Avatar

Min-Chuan Huang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ji-Shiang Hung

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chi-Hau Chen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chih-Hsing Chou

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Hong-Shiee Lai

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chiung-Hui Liu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Rey-Heng Hu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Yao-Ming Wu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chia-Hua Chen

National Taiwan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge