Network


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

Hotspot


Dive into the research topics where Sheng-Ming Wu is active.

Publication


Featured researches published by Sheng-Ming Wu.


Oncogene | 2011

Cathepsin H regulated by the thyroid hormone receptors associate with tumor invasion in human hepatoma cells.

Sheng-Ming Wu; Ya Hui Huang; Chau-Ting Yeh; Ming-Ming Tsai; Chia-Jung Liao; Wan Li Cheng; Wei-Jan Chen; Kwang-Huei Lin

Thyroid hormone, 3, 3′, 5-triiodo-L-thyronine (T3), mediates cell growth, development and differentiation by binding to its nuclear receptors (TRs). The role of TRs in cancer is still undefined. Notably, hyperthyroxinemia has been reported to influence the rate of colon cancer in an experimental model of carcinogenesis in rats. Previous microarray analysis revealed that cathepsin H (CTSH) is upregulated by T3 in HepG2-TR cells. We verified that mRNA and protein expression of CTSH are induced by T3 in HepG2-TR cells and in thyroidectomized rats following administration of T3. The possible thyroid hormone-responsive elements of the CTSH promoter localized to the nucleotides –2038 to –1966 and –1565 to –1501 regions. An in vitro functional assay showed that CTSH can increase metastasis. J7 cells overexpressing CTSH were inoculated into severe combined immune-deficient mice and these J7-CTSH mice displayed a greater metastatic potential than did J7-control mice. The clinicopathologic significance of CTSH expression in hepatocellular carcinoma (HCC) was also investigated. The CTSH overexpressing in HCC was associated with the presence of microvascular invasion (P=0.037). The microvascular invasion characteristic is closely related to our in vitro characterization of CTSH function. Our results show that T3-mediated upregulation of CTSH led to matrix metallopeptidase or extracellular signal-regulated kinase activation and increased cell migration. This study demonstrated that CTSH overexpression in a subset hepatoma may be TR dependent and suggests that this overexpression has an important role in hepatoma progression.


Hepatology | 2012

Dickkopf 4 positively regulated by the thyroid hormone receptor suppresses cell invasion in human hepatoma cells.

Chen-Hsin Liao; Chau-Ting Yeh; Ya-Hui Huang; Sheng-Ming Wu; Hsiang-Cheng Chi; Ming-Ming Tsai; Chung-Ying Tsai; Chia-Jung Liao; Yi-Hsin Tseng; Yang-Hsiang Lin; Cheng-Yi Chen; I-Hsiao Chung; Wan-Li Cheng; Wei-Jan Chen; Kwang-Huei Lin

Thyroid hormone (T3) mediates cellular growth, development, and differentiation by binding to the nuclear thyroid hormone receptor (TR). Recent studies suggest that long‐term hypothyroidism is associated with human hepatocellular carcinoma (HCC) independent from other major HCC risk factors. Dickkopf (DKK) 4, a secreted protein, antagonizes the Wnt signal pathway. In this study, we demonstrate that T3 may play a suppressor role by inducing DKK4 expression in HCC cells at both the messenger RNA (mRNA) and protein levels. DKK4 was down‐regulated in 67.5% of HCC cancerous tissues. The decrease in DKK4 levels was accompanied by a concomitant decrease in TR protein levels in the matched cancerous tissues in 31% of tissues compared by immunoblotting with the adjacent noncancerous tissues. Further, TR and DKK4 expression levels were positively correlated in both normal and cancerous specimens by tissue array analysis. In function assays, stable DKK4 transfected into J7 or HepG2 cells decreased cell invasion in vitro. Conversely, knocking down DKK4 restores cell invasiveness. DKK4‐expressing J7 clones showed increased degradation of β‐catenin, but down‐regulation of CD44, cyclin D1, and c‐Jun. To investigate the effect of DKK4 and TR on tumor growth in vivo, we established a xenograft of J7 cells in nude mice. J7‐DKK4 and J7‐TRα1 overexpressing mice, which displayed growth arrest, lower lung colony formation index, and smaller tumor size than in control mice, supporting an inhibitory role of DKK4 in tumor progression. Conclusion: Taken together, these data suggest that the TR/DKK4/Wnt/β‐catenin cascade influences the proliferation and migration of hepatoma cells during the metastasis process and support a tumor suppressor role of the TR. (Hepatology 2012)


Oncogene | 2013

Thyroid hormone receptor represses miR-17 expression to enhance tumor metastasis in human hepatoma cells

Yang-Hsiang Lin; Chia Jung Liao; Ya Hui Huang; Meng-Han Wu; Hsiang-Cheng Chi; Sheng-Ming Wu; Chun-Chieh Chen; Yi-Hsin Tseng; Chung-Ying Tsai; I-Hsiao Chung; Tzu-I Wu; Ming-Ming Tsai; Crystal D. Lin; Kwang-Huei Lin

MicroRNAs (miRNAs) are thought to control tumor metastasis through direct interactions with target genes. Thyroid hormone (T3) and its receptor (TR) are involved in cell growth and cancer progression. However, the issue of whether miRNAs participate in T3/TR-mediated tumor migration is yet to be established. In the current study, we demonstrated that T3/TR negatively regulates mature miR-17 transcript expression, both in vitro and in vivo. Luciferase reporter and chromatin immunoprecipitation (ChIP) assays localized the regions responding to TR-mediated repression to positions −2234/−2000 of the miR-17 promoter sequence. Overexpression of miR-17 markedly inhibited cell migration and invasion in vitro and in vivo, mediated via suppression of matrix metalloproteinases (MMP)-3. Moreover, p-AKT expression was increased in miR-17-knockdown cells that led to enhanced cell invasion, which was blocked by LY294002. Notably, low miR-17 expression was evident in highly metastatic cells. The cell migration ability was increased by T3, but partially reduced upon miR-17 overexpression. Notably, TRα1 was frequently upregulated in hepatocellular carcinoma (HCC) samples and associated with low overall survival (P=0.023). miR-17 expression was significantly negatively associated with TRα1 (P=0.033) and MMP3 (P=0.043) in HCC specimens. Data from our study suggest that T3/TR, miR-17, p-AKT and MMP3 activities are interlinked in the regulation of cancer cell metastasis.


PLOS ONE | 2012

Glyoxalase-I is a novel prognosis factor associated with gastric cancer progression.

Wan-Li Cheng; Ming-Ming Tsai; Chung-Ying Tsai; Ya-Hui Huang; Cheng-Yi Chen; Hsiang Cheng Chi; Yi-Hsin Tseng; Im-Wai Chao; Wei-Chi Lin; Sheng-Ming Wu; Ying Liang; Chia-Jung Liao; Yang-Hsiang Lin; I-Hsiao Chung; Wei-Jan Chen; Paul Y. Lin; Chia-Siu Wang; Kwang-Huei Lin

Glyoxalase I (GLO1), a methylglyoxal detoxification enzyme, is implicated in the progression of human malignancies. The role of GLO1 in gastric cancer development or progression is currently unclear. The expression of GLO1 was determined in primary gastric cancer specimens using quantitative polymerase chain reaction, immunohistochemistry (IHC), and western blotting analyses. GLO1 expression was higher in gastric cancer tissues, compared with that in adjacent noncancerous tissues. Elevated expression of GLO1 was significantly associated with gastric wall invasion, lymph node metastasis, and pathological stage, suggesting a novel role of GLO1 in gastric cancer development and progression. The 5-year survival rate of the lower GLO1 expression groups was significantly greater than that of the higher expression groups (log rank P = 0.0373) in IHC experiments. Over-expression of GLO1 in gastric cancer cell lines increases cell proliferation, migration and invasiveness. Conversely, down-regulation of GLO1 with shRNA led to a marked reduction in the migration and invasion abilities. Our data strongly suggest that high expression of GLO1 in gastric cancer enhances the metastasis ability of tumor cells in vitro and in vivo, and support its efficacy as a potential marker for the detection and prognosis of gastric cancer.


Cell Death & Differentiation | 2012

Thyroid hormone receptors promote metastasis of human hepatoma cells via regulation of TRAIL

Hsiang-Cheng Chi; Shen Liang Chen; Chia Jung Liao; Chia-Jung Liao; Tsai Mm; Yang-Hsiang Lin; Yin-Cheng Huang; Chau-Ting Yeh; Sheng-Ming Wu; Yi-Hsin Tseng; Chun-Chieh Chen; Chung-Ying Tsai; I-Hsiao Chung; Wei-Jan Chen; Kwang-Huei Lin

Although accumulating evidence has confirmed the important roles of thyroid hormone (T3) and its receptors (TRs) in tumor progression, the specific functions of TRs in carcinogenesis remain unclear. In the present study, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) was directly upregulated by T3 in TR-overexpressing hepatoma cell lines. TRAIL is an apoptotic inducer, but it can nonetheless trigger non-apoptotic signals favoring tumorigenesis in apoptosis-resistant cancer cells. We found that TR-overexpressing hepatoma cells treated with T3 were apoptosis resistant, even when TRAIL was upregulated. This apoptotic resistance may be attributable to simultaneous upregulation of Bcl-xL by T3, because (1) knockdown of T3-induced Bcl-xL expression suppressed T3-mediated protection against apoptosis, and (2) overexpression of Bcl-xL further protected hepatoma cells from TRAIL-induced apoptotic death, consequently leading to TRAIL-promoted metastasis of hepatoma cells. Moreover, T3-enhanced metastasis in vivo was repressed by the treatment of TRAIL-blocking antibody. Notably, TRAIL was highly expressed in a subset of hepatocellular carcinoma (HCC) patients, and this high-level expression was significantly correlated with that of TRs in these HCC tissues. Together, our findings provide evidence for the existence of a novel mechanistic link between increased TR and TRAIL levels in HCC. Thus, TRs induce TRAIL expression, and TRAIL thus synthesized acts in concert with simultaneously synthesized Bcl-xL to promote metastasis, but not apoptosis.


Cellular and Molecular Life Sciences | 2010

Thyroid hormone receptor-mediated regulation of the methionine adenosyltransferase 1 gene is associated with cell invasion in hepatoma cell lines

Sheng-Ming Wu; Ya-Hui Huang; Yi-Hsin Lu; Ling-Fang Chien; Chau-Ting Yeh; Ming-Ming Tsai; Chen-Hsin Liao; Wei-Jan Chen; Chia-Jung Liao; Wan-Li Cheng; Kwang-Huei Lin

The thyroid hormone T3 regulates differentiation, growth, and development. We demonstrated that methionine adenosyltransferase 1A (MAT1A) was positively regulated by T3 identified by cDNA microarray previously. The expression of the MAT1A was upregulated by T3 in hepatoma cell lines overexpressing thyroid hormone receptors (TRs). Additionally, these findings indicate that MAT1A may be regulated by CCAAT/enhancer binding protein (C/EBP). The critical role of the C/EBP binding sites was confirmed by the reporter or chromatin immuno-precipitation (ChIP) assay. In addition, C/EBP was upregulated in hepatoma cells after T3 treatment and ectopic expression of MAT1A inhibited cell migration and invasion in J7 hepatoma cells. Conversely, knockdown of MAT1A expression increased cell migration. Together, these findings suggest that the expression of the MAT1A gene is mediated by C/EBP and is indirectly upregulated by T3. Finally, TR was downregulated in a small subset of hepatocellular carcinoma cells concomitantly reduced the expression of C/EBPα and MAT1A.


Journal of Hepatology | 2015

Repression of microRNA-130b by thyroid hormone enhances cell motility

Yang-Hsiang Lin; Meng-Han Wu; Chia-Jung Liao; Ya-Hui Huang; Hsiang-Cheng Chi; Sheng-Ming Wu; Cheng-Yi Chen; Yi-Hsin Tseng; Chung-Ying Tsai; I-Hsiao Chung; Ming-Ming Tsai; Ching-Ying Chen; Tina P. Lin; Yung-Hsin Yeh; Wei-Jan Chen; Kwang-Huei Lin

BACKGROUND & AIMS Thyroid hormone (T3) and its receptor (TR) are involved in cell growth and cancer progression. Although deregulation of microRNA (miRNA) expression has been detected in many tumor types, the mechanisms underlying functional impairment and specific involvement of miRNAs in tumor metastasis remain unclear. In the current study, we aimed to elucidate the involvement of deregulated miRNA-130b (miR-130b) and its target genes mediated by T3/TR in cancer progression. METHODS Quantitative reverse transcription-PCR, luciferase and chromatin immunoprecipitation assays were performed to identify the miR-130b transcript and the mechanisms implicated in its regulation. The effects of miR-130b on hepatocellular carcinoma (HCC) invasion were further examined in vitro and in vivo. Clinical correlations among miR-130b, TRs and interferon regulatory factor 1 (IRF1) were examined in HCC samples using Spearman correlation analysis. RESULTS Our experiments disclosed negative regulation of miR-130b expression by T3/TR. Overexpression of miR-130b led to marked inhibition of cell migration and invasion, which was mediated via suppression of IRF1. Cell migration ability was promoted by T3, but partially suppressed upon miR-130b overexpression. Furthermore, miR-130b suppressed expression of epithelial-mesenchymal transition (EMT)-related genes, matrix metalloproteinase-9, phosphorylated mammalian target of rapamycin (mTOR), p-ERK1/2, p-AKT and p-signal transducer and activator of transcription (STAT)-3. Notably, miR-130b was downregulated in hepatoma samples and its expression patterns were inversely correlated with those of TRα1 and IRF1. CONCLUSIONS Our data collectively highlight a novel pathway interlinking T3/TR, miR-130b, IRF1, the EMT-related genes, p-mTOR, p-STAT3 and the p-AKT cascade, which regulates the motility and invasion of hepatoma cells.


Cancer Science | 2012

Overexpression of ADP-ribosylation factor 1 in human gastric carcinoma and its clinicopathological significance

Ming-Ming Tsai; Paul Y. Lin; Wan-Li Cheng; Chung-Ying Tsai; Hsiang-Cheng Chi; Cheng-Yi Chen; Yi-Hsin Tseng; Yi-Fen Cheng; Chi-De Chen; Ying Liang; Chia-Jung Liao; Sheng-Ming Wu; Yang-Hsiang Lin; I-Hsiao Chung; Chia-Siu Wang; Kwang-Huei Lin

Gastric cancer is the sixth leading cause of cancer‐related death in Taiwan, and the identification of related factors is essential to increase patient survival. ADP‐ribosylation factor 1 (ARF1) was initially identified using 2‐D electrophoresis combined with MALDI–time‐of‐flight mass spectrometry. ADP‐ribosylation factor 1 belongs to the Ras superfamily or GTP‐binding protein family and has been shown to enhance cell proliferation. In the current study, we evaluated the potential of ARF1 as a biomarker for gastric cancer detection. ADP‐ribosylation factor 1 mRNA was upregulated in tumor tissues (compared with adjacent non‐tumor tissues, n = 55) in approximately 67.2% of gastric cancer patients. Expression of ARF1 protein was additionally observed using Western blot and immunohistochemistry (IHC) analyses. The clinicopathological correlations of ARF1 were further evaluated. Elevated ARF1 expression was strongly correlated with lymph node metastasis (P = 0.008), serosal invasion (P = 0.046), lymphatic invasion (P = 0.035), and pathological staging (P = 0.010). Moreover, the 5‐year survival rate for the lower ARF1 expression group (n = 50; IHC score < 90) was higher than that of the higher expression group (n = 60; IHC score ≥ 90) (P = 0.0228, log–rank test). To establish the specific function of ARF1 in human gastric cancer, isogenic ARF1‐overexpressing cell lines were prepared. Our results showed that ARF1‐overexpressing clones display enhanced cell proliferation, migration, and invasion. Furthermore, ARF1‐overexpression might contribute to poor prognosis of patients. These findings collectively support the utility of ARF1 as a novel prognostic marker for gastric cancer and its role in cell invasion. Cancer Sci 2012; 103: 1136–1144)


Oncogene | 2013

Thyroid hormone suppresses cell proliferation through endoglin-mediated promotion of p21 stability

Yang-Hsiang Lin; Yin-Cheng Huang; Meng-Han Wu; Sheng-Ming Wu; Hsiang-Cheng Chi; Chia Jung Liao; Chun-Chieh Chen; Yi-Hsin Tseng; Chung-Ying Tsai; Tsai Mm; Kwang-Huei Lin

Hypothyroidism has been associated with significantly elevated risk for hepatocellular carcinoma (HCC), although the precise underlying mechanisms remain unknown at present. Thyroid hormone (T3) and its receptor (TR) are involved in metabolism and growth. Endoglin is a T3/TR candidate target gene identified from our previous studies. Here, we demonstrated that T3 positively regulates endoglin mRNA and protein levels, both in vitro and in vivo. The thyroid hormone response elements of endoglin were identified at positions −2114/−2004 and −2032/−1973 of the promoter region using the electrophoretic mobility shift assay and chromatin immunoprecipitation assay. Endoglin was downregulated in the subgroups of HCC patients and significantly associated with histology grade (negative association, P=0.001), and this expression level was significantly associated with TRα1 in these HCC patients. Our results clearly indicate that p21 is involved in T3-mediated suppression of cell proliferation. Knock down of endoglin expression in HCC cells facilitated p21 polyubiquitination and promoted cell proliferation in the presence of T3. The data collectively suggest that T3/TR signaling suppresses cell proliferation by upregulating endoglin, in turn, affecting p21 stability. The results indicate that endoglin has a suppressor role to inhibit cell proliferation in HCC cell lines.


Biochemical and Biophysical Research Communications | 2013

Thyroid hormone receptor inhibits hepatoma cell migration through transcriptional activation of Dickkopf 4.

Hsiang-Cheng Chi; Chen-Hsin Liao; Ya-Hui Huang; Sheng-Ming Wu; Chung-Ying Tsai; Chia-Jung Liao; Yi-Hsin Tseng; Yang-Hsiang Lin; Cheng-Yi Chen; I-Hsiao Chung; Tzu-I Wu; Wei-Jan Chen; Kwang-Huei Lin

Collaboration


Dive into the Sheng-Ming Wu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ming-Ming Tsai

Chang Gung University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge