Network


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

Hotspot


Dive into the research topics where Shine Gwo Shiah is active.

Publication


Featured researches published by Shine Gwo Shiah.


Cancer Research | 2010

H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM

Min Wei Chen; Kuo-Tai Hua; Hsin Jung Kao; Chia Chun Chi; Lin Hung Wei; Gunnar Johansson; Shine Gwo Shiah; Pai Sheng Chen; Yung-Ming Jeng; Tsu-Yao Cheng; Tsung Ching Lai; Jeng Shou Chang; Yi Hua Jan; Ming Hsien Chien; Chih Jen Yang; Ming Shyan Huang; Michael Hsiao; Min-Liang Kuo

G9a is a mammalian histone methyltransferase that contributes to the epigenetic silencing of tumor suppressor genes. Emerging evidence suggests that G9a is required to maintain the malignant phenotype, but the role of G9a function in mediating tumor metastasis has not been explored. Here, we show that G9a is expressed in aggressive lung cancer cells, and its elevated expression correlates with poor prognosis. RNAi-mediated knockdown of G9a in highly invasive lung cancer cells inhibited cell migration and invasion in vitro and metastasis in vivo. Conversely, ectopic G9a expression in weakly invasive lung cancer cells increased motility and metastasis. Mechanistic investigations suggested that repression of the cell adhesion molecule Ep-CAM mediated the effects of G9a. First, RNAi-mediated knockdown of Ep-CAM partially relieved metastasis suppression imposed by G9a suppression. Second, an inverse correlation between G9a and Ep-CAM expression existed in primary lung cancer. Third, Ep-CAM repression was associated with promoter methylation and an enrichment for dimethylated histone H3K9. G9a knockdown reduced the levels of H3K9 dimethylation and decreased the recruitment of the transcriptional cofactors HP1, DNMT1, and HDAC1 to the Ep-CAM promoter. Our findings establish a functional contribution of G9a overexpression with concomitant dysregulation of epigenetic pathways in lung cancer progression.


Journal of Cellular Biochemistry | 1999

Nitric oxide prevents apoptosis of human endothelial cells from high glucose exposure during early stage

Feng M. Ho; Shing H. Liu; Chiau S. Liau; Por J. Huang; Shine Gwo Shiah; Shoei Yn Lin-Shiau

Hyperglycemia is a major cause of diabetic vascular disease. High glucose can induce reactive oxygen species (ROS) and nitric oxide (NO) generation, which can subsequently induce endothelial dysfunction. High glucose is also capable of triggering endothelial cell apoptosis. Little is known about the molecular mechanisms and the role of ROS and NO in high glucose‐induced endothelial cell apoptosis. This study was designed to determine the involvement of ROS and NO in high glucose‐induced endothelial cell apoptosis. Expression of endothelial nitric oxide synthase (eNOS) protein and apoptosis were studied in cultured human umbilical vein endothelial cells (HUVECs) exposed to control‐level (5.5 mM) and high‐level (33 mM) glucose at various periods (e.g., 2, 12, 24, 48 h). We also examined the effect of high glucose on H2O2 production using flow cytometry. The results showed that eNOS protein expression was up‐regulated by high glucose exposure for 2–6 h and gradually reduced after longer exposure in HUVECs. H2O2 production and apoptosis, which can be reversed by vitamin C and NO donor (sodium nitroprusside), but enhanced by NOS inhibitor (NG‐nitro‐L‐arginine methyl ether), were collated to a different time course (24–48 h) to HUVECs. These results provide the molecular basis for understanding that NO plays a protective role from apoptosis of HUVECs during the early stage (<24 h) of high glucose exposure, but in the late stage (>24 h), high glucose exposure leads to the imbalance of NO and ROS, resulting to the observed apoptosis. This may explain, at least in part, the impaired endothelial function and vascular complication of diabetic mellitus that would occur at late stages. J. Cell. Biochem. 75:258–263, 1999.


Biochimica et Biophysica Acta | 2013

Nutrient deprivation induces the Warburg effect through ROS/AMPK-dependent activation of pyruvate dehydrogenase kinase

Ching An Wu; Yee Chao; Shine Gwo Shiah; Wan-Wan Lin

The Warburg effect is known to be crucial for cancer cells to acquire energy. Nutrient deficiencies are an important phenomenon in solid tumors, but the effect on cancer cell metabolism is not yet clear. In this study, we demonstrate that starvation of HeLa cells by incubation with Hanks buffered salt solution (HBSS) induced cell apoptosis, which was accompanied by the induction of reactive oxygen species (ROS) production and AMP-activated protein kinase (AMPK) phosphorylation. Notably, HBSS starvation increased lactate production, cytoplasmic pyruvate content and decreased oxygen consumption, but failed to change the lactate dehydrogenase (LDH) activity or the glucose uptake. We found that HBSS starvation rapidly induced pyruvate dehydrogenase kinase (PDK) activation and pyruvate dehydrogenase (PDH) phosphorylation, both of which were inhibited by compound C (an AMPK inhibitor), NAC (a ROS scavenger), and the dominant negative mutant of AMPK. Our data further revealed the involvement of ROS production in AMPK activation. Moreover, DCA (a PDK inhibitor), NAC, and compound C all significantly decreased HBSS starvation-induced lactate production accompanied by enhancement of HBSS starvation-induced cell apoptosis. Not only in HeLa cells, HBSS-induced lactate production and PDH phosphorylation were also observed in CL1.5, A431 and human umbilical vein endothelial cells. Taken together, we for the first time demonstrated that a low-nutrient condition drives cancer cells to utilize glycolysis to produce ATP, and this increases the Warburg effect through a novel mechanism involving ROS/AMPK-dependent activation of PDK. Such an event contributes to protecting cells from apoptosis upon nutrient deprivation.


Molecular Cancer | 2014

Reciprocal regulation of MicroRNA-99a and insulin-like growth factor I receptor signaling in oral squamous cell carcinoma cells

Yi Chen Yen; Shine Gwo Shiah; Hsiao Chien Chu; Yuan Ming Hsu; Jenn Ren Hsiao; Jang Yang Chang; Wen Chun Hung; Chun Ta Liao; Ann-Joy Cheng; Ya Ching Lu; Ya Wen Chen

BackgroundMicroRNAs (miRNAs), small noncoding RNA molecules can function as oncogenes or tumor suppressors in tumorigenesis. Oral squamous cell carcinoma (OSCC) is one of the most prevalent cancers worldwide with a 5-year survival rate of approximately 50%.MethodsThe expression of microRNA-99a (miR-99a) in OSCC tissues and cell lines was investigated using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis. The functions of miR-99a in migration/invasion and lung colonization were determined by transwell and tail vein injection assays, respectively. Specific targets of miR-99a were determined by software prediction, correlation with target protein expression, and luciferase reporter assay. The signaling pathways involved in regulation of miR-99a were investigated using the kinase inhibitors.ResultsWe observed reduced levels of miR-99a, identified as one of the most downregulated miRNA in OSCC and all tested OSCC cell lines compared to normal oral keratinocytes. Ectopic miR-99a expression in OSCC cells markedly reduced migration and invasion in vitro as well as lung colonization in vivo. When evaluating the specific targets of miR-99a, we found that ectopic miR-99a expression downregulates insulin-like growth factor 1 receptor (IGF1R) protein and that the expression of miR-99a correlates negatively with IGF1R protein in OSCC cells. Insertion of the 3′UTR of IGF1R mRNA into the 3′UTR of a reporter gene markedly reduced luciferase activity in OSCC cells expressing miR-99a, suggesting that miR-99a reduces luciferase activity by targeting the 3′UTR of IGF1R mRNA. When evaluating the mechanisms of miR-99a downregulation, we observed the upregulation of miR-99a expression in serum-starved conditions and its suppression in response to insulin-like growth factor (IGF1) stimulation. Inhibitors of phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) kinase inhibited IGF1-induced suppression of miR-99a, suggesting the negative regulation of miR-99a expression by IGF1R signaling.ConclusionOverall, results indicate that miR-99a functions as a tumor metastasis suppressor in OSCC cells and mutually regulates IGF1R expression in a reciprocal regulation.


The Journal of Pathology | 2016

Up‐regulation of miR‐455‐5p by the TGF‐β–SMAD signalling axis promotes the proliferation of oral squamous cancer cells by targeting UBE2B

Chao-Min Cheng; Shine Gwo Shiah; Chien Chang Huang; Jenn Ren Hsiao; Jang Yang Chang

MicroRNAs (miRNAs) are involved in the tumourigenesis of various cancers by regulating their downstream targets. To identify the changes of miRNAs in oral squamous cell carcinoma (OSCC), we investigated the expression profiles of miRNAs in 40 pairs of OSCC specimens and their matched non‐tumour epithelial tissues. Our data revealed higher miR‐455‐5p expression in the tumour tissues than in the normal tissues; the expression was also higher in oral cancer cell lines than in normal keratinocyte cell lines. MiR‐455‐5p knockdown reduced both the anchorage‐independent growth and the proliferative ability of oral cancer cells, and these factors increased in miR‐455‐5p‐overexpressing cells. Furthermore, by analysing the array data of patients with cancer and cell lines, we identified ubiquitin‐conjugating enzyme E2B (UBE2B) as a target of miR‐455‐5p, and further validated this using 3′‐untranslated region luciferase reporter assays and western blot analysis. We also demonstrated that UBE2B suppression rescued the impaired growth ability of miR‐455‐5p‐knockdown cells. Furthermore, we observed that miR‐455‐5p expression was regulated, at least in part, by the transforming growth factor‐β (TGF‐β) pathway through the binding of SMAD3 to specific promoter regions. Notably, miR‐455‐5p expression was associated with the nodal status, stage, and overall survival in our patients, suggesting that miR‐455‐5p is a potential marker for predicting the prognosis of patients with oral cancer. In conclusion, we reveal that miR‐455‐5p expression is regulated by the TGF‐β‐dependent pathway, which subsequently leads to UBE2B down‐regulation and contributes to oral cancer tumourigenesis. Copyright


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.


PLOS ONE | 2015

Elevation of Soluble Guanylate Cyclase Suppresses Proliferation and Survival of Human Breast Cancer Cells

Hui Chin Wen; Chih Pin Chuu; Chen Yu Chen; Shine Gwo Shiah; Hsing Jien Kung; Kuang Liang King; Liang Chen Su; Shi-Chuan Chang; Chung Ho Chang

Nitric oxide (NO) is an essential signaling molecule in biological systems. Soluble guanylate cyclase (sGC), composing of α1 and β1 subunit, is the receptor for NO. Using radioimmunoassay, we discovered that activation of sGC by treatment with bradykinin or sodium nitroprusside (SNP) is impaired in MCF-7 and MDA-MB-231 breast cancer cells as compared to normal breast epithelial 184A1 cells. The 184A1 cells expressed both sGC α1 and sGCβ1 mRNAs. However, levels of sGCβ1 mRNAs were relatively lower in MCF-7 cells while both mRNA of sGC subunits were absent in MDA-MB-231 cells. Treatment with DNA methyltransferase inhibitor 5-aza-2’-deoxycytidine (5-aza-dC) increased mRNA levels of both sGCα1 and sGCβ1 in MDA-MB-231 cells but only sGCβ1 mRNAs in MCF-7 cells. The 5-aza-dC treatment increased the SNP-induced cGMP production in MCF-7 and MDA-MB-231, but not in 184A1 cells. Bisulfite sequencing revealed that the promoter of sGCα1 in MDA-MB-231 cells and promoter of sGCβ1 in MCF-7 cells were methylated. Promoter hypermethylation of sGCα1 and sGCβ1 was found in 1 out of 10 breast cancer patients. Over-expression of both sGC subunits in MDA-MB-231 cells induced apoptosis and growth inhibition in vitro as well as reduced tumor incidence and tumor growth rate of MDA-MB-231 xenografts in nude mice. Elevation of sGC reduced protein abundance of Bcl-2, Bcl-xL, Cdc2, Cdc25A, Cyclin B1, Cyclin D1, Cdk6, c-Myc, and Skp2 while increased protein expression of p53. Our study demonstrated that down-regulation of sGC, partially due to promoter methylation, provides growth and survival advantage in human breast cancer cells.


Molecular Oncology | 2016

IL-8 induces miR-424-5p expression and modulates SOCS2/ STAT5 signaling pathway in oral squamous cell carcinoma

Hsuan Yu Peng; Shih Sheng Jiang; Jenn Ren Hsiao; Michael Hsiao; Yuan Ming Hsu; Guan Hsun Wu; Wei Min Chang; Jang Yang Chang; Shiow Lian Catherine Jin; Shine Gwo Shiah

Suppressor of cytokine signaling (SOCS) proteins are negative feedback regulators of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway. Dysregulation of SOCS protein expression in cancers can be one of the mechanisms that maintain STAT activation, but this mechanism is still poorly understood in oral squamous cell carcinoma (OSCC). Here, we report that SOCS2 protein is significantly downregulated in OSCC patients and its levels are inversely correlated with miR‐424‐5p expression. We identified the SOCS2 protein, which modulates STAT5 activity, as a direct target of miR‐424‐5p. The miR‐424‐5p‐induced STAT5 phosphorylation, matrix metalloproteinases (MMPs) expression, and cell migration and invasion were blocked by SOCS2 restoration, suggesting that miR‐424‐5p exhibits its oncogenic activity through negatively regulating SOCS2 levels. Furthermore, miR‐424‐5p expression could be induced by the cytokine IL‐8 primarily through enhancing STAT5 transcriptional activity rather than NF‐κB signaling. Antagomir‐mediated inactivation of miR‐424‐5p prevented the IL‐8‐induced cell migration and invasion, indicating that miR‐424‐5p is required for IL‐8‐induced cellular invasiveness. Taken together, these data indicate that STAT5‐dependent expression of miR‐424‐5p plays an important role in mediating IL‐8/STAT5/SOCS2 feedback loop, and scavenging miR‐424‐5p function using antagomir may have therapeutic potential for the treatment of OSCC.


PLOS ONE | 2013

Regulation of c-Fos Gene Expression by NF-κB: A p65 Homodimer Binding Site in Mouse Embryonic Fibroblasts but Not Human HEK293 Cells

Yu Cheng Tu; Duen Yi Huang; Shine Gwo Shiah; Jang Shiun Wang; Wan-Wan Lin

The immediate early gene c-Fos is reported to be regulated by Elk-1 and cAMP response element-binding protein (CREB), but whether nuclear factor (NF)-κB is also required for controlling c-Fos expression is unclear. In this study, we determined how NF-κB’s coordination with Elk/serum response factor (SRF) regulates c-fos transcription. We report that PMA strongly induced c-Fos expression, but tumor necrosis factor (TNF)-α did not. In mouse embryonic fibroblasts, the PMA induction of c-Fos was suppressed by a deficiency in IKKα, IKKβ, IKKγ, or p65. By contrast, in human embryonic kidney 293 cells, PMA induced c-Fos independently of p65. In accordance with these results, we identified an NF-κB binding site in the mouse but not human c-fos promoter. Under PMA stimulation, IKKα/β mediated p65 phosphorylation and the binding of the p65 homodimer to the NF-κB site in the mouse c-fos promoter. Furthermore, our studies demonstrated independent but coordinated functions of the IKKα/β-p65 and extracellular signal-regulated kinase (ERK)-Elk-1 pathways in the PMA induction of c-Fos. Collectively, these results reveal the distinct requirement of NF-κB for mouse and human c-fos regulation. Binding of the p65 homodimer to the κB site was indispensable for mouse c-fos expression, whereas the κB binding site was not present in the human c-fos promoter. Because of an inability to evoke sufficient ERK activation and Elk-1 phosphorylation, TNF-α induces c-Fos more weakly than PMA does in both mouse and human cells.


PLOS ONE | 2016

MPT0B098, a microtubule inhibitor, suppresses JAK2/STAT3 signaling pathway through modulation of SOCS3 stability in oral squamous cell carcinoma

Hsuan Yu Peng; Yun Ching Cheng; Yuan Ming Hsu; Guan Hsun Wu; Ching Chuan Kuo; Jing Ping Liou; Jang Yang Chang; Shiow Lian Catherine Jin; Shine Gwo Shiah

Microtubule inhibitors have been shown to inhibit Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signal transduction pathway in various cancer cells. However, little is known of the mechanism by which the microtubule inhibitors inhibit STAT3 activity. In the present study, we examined the effect of a novel small-molecule microtubule inhibitor, MPT0B098, on STAT3 signaling in oral squamous cell carcinoma (OSCC). Treatment of various OSCC cells with MPT0B098 induced growth inhibition, cell cycle arrest and apoptosis, as well as increased the protein level of SOCS3. The accumulation of SOCS3 protein enhanced its binding to JAK2 and TYK2 which facilitated the ubiquitination and degradation of JAK2 and TYK2, resulting in a loss of STAT3 activity. The inhibition of STAT3 activity led to sensitization of OSCC cells to MPT0B098 cytotoxicity, indicating that STAT3 is a key mediator of drug resistance in oral carcinogenesis. Moreover, the combination of MPT0B098 with the clinical drug cisplatin or 5-FU significantly augmented growth inhibition and apoptosis in OSCC cells. Taken together, our results provide a novel mechanism for the action of MPT0B098 in which the JAK2/STAT3 signaling pathway is suppressed through the modulation of SOCS3 protein level. The findings also provide a promising combinational therapy of MPT0B098 for OSCC.

Collaboration


Dive into the Shine Gwo Shiah's collaboration.

Top Co-Authors

Avatar

Jang Yang Chang

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Jenn Ren Hsiao

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hsuan Yu Peng

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Yi Shing Shieh

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yuan Ming Hsu

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Guan Hsun Wu

National Health Research Institutes

View shared research outputs
Top Co-Authors

Avatar

Min-Liang Kuo

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chi Long Chen

Taipei Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge