Network


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

Hotspot


Dive into the research topics where Yu-Jen Wu is active.

Publication


Featured researches published by Yu-Jen Wu.


International Journal of Molecular Sciences | 2013

Inhibition of Melanogenesis by Gallic Acid: Possible Involvement of the PI3K/Akt, MEK/ERK and Wnt/β-Catenin Signaling Pathways in B16F10 Cells

Tzu-Rong Su; Jen-Jie Lin; Chi-Chu Tsai; Tsu-Kei Huang; Zih-Yan Yang; Ming-O Wu; Yu-Qing Zheng; Ching-Chyuan Su; Yu-Jen Wu

Gallic acid is one of the major flavonoids found in plants. It acts as an antioxidant, and seems to have anti-inflammatory, anti-viral, and anti-cancer properties. In this study, we investigated the effects of gallic acid on melanogenesis, including the activation of melanogenesis signaling pathways. Gallic acid significantly inhibited both melanin synthesis and tyrosinase activity in a dose- and time-dependent manner, and decreased the expression of melanogenesis-related proteins, such as microphthalmia-associated transcription factor (MITF), tyrosinase, tyrosinase-related protein-1 (TRP1), and dopachrome tautomerase (Dct). In addition, gallic acid also acts by phosphorylating and activating melanogenesis inhibitory proteins such as Akt and mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK). Using inhibitors against PI3K/Akt (LY294002) or MEK/ERK-specific (PD98059), the hypopigmentation effect was suppressed, and the gallic acid-initiated activation of MEK/ERK and PI3K/Akt was also revoked. Gallic acid also increased GSK3β and p-β-catenin expression but down-regulated p-GSK3β. Moreover, GSK3β-specific inhibitor (SB216763) restored gallic acid-induced melanin reduction. These results suggest that activation of the MEK/ERK, PI3K/Akt, and inhibition of Wnt/β-catenin signaling pathways is involved in the melanogenesis signaling cascade, and that activation by gallic acid reduces melanin synthesis via down-regulation of MITF and its downstream signaling pathway. In conclusion, gallic acid may be a potentially agent for the treatment of certain skin conditions.


Marine Drugs | 2011

Proteomic Analysis of Anti-Tumor Effects of 11-Dehydrosinulariolide on CAL-27 Cells

Chih-I Liu; Cheng-Chi Chen; Jiing-Chuan Chen; Jui-Hsin Su; Han Hsiang Huang; Jeff Yi-Fu Chen; Yu-Jen Wu

The anti-tumor effects of 11-dehydrosinulariolide, an active ingredient isolated from soft coral Sinularia leptoclados, on CAL-27 cells were investigated in this study. In the MTT assay for cell proliferation, increasing concentrations of 11-dehydrosinulariolide decreased CAL-27 cell viability. When a concentration of 1.5 μg/mL of 11-dehydrosinulariolide was applied, the CAL-27 cells viability was reduced to a level of 70% of the control sample. The wound healing function decreased as the concentration of 11-dehydrosinulariolide increased. The results in this study indicated that treatment with 11-dehydrosinulariolide for 6 h significantly induced both early and late apoptosis of CAL-27 cells, observed by flow cytometric measurement and microscopic fluorescent observation. A comparative proteomic analysis was conducted to investigate the effects of 11-dehydrosinulariolide on CAL-27 cells at the molecular level by comparison between the protein profiling (revealed on a 2-DE map) of CAL-27 cells treated with 11-dehydrosinulariolide and that of CAL-27 cells without the treatment. A total of 28 differential proteins (12 up-regulated and 16 down-regulated) in CAL-27 cells treated with 11-dehydrosinulariolide have been identified by LC-MS/MS analysis. Some of the differential proteins are associated with cell proliferation, apoptosis, protein synthesis, protein folding, and energy metabolism. The results of this study provided clues for the investigation of biochemical mechanisms of the anti-tumor effects of 11-dehydrosinulariolide on CAL-27 cells and could be valuable information for drug development and progression monitoring of oral squamous cell carcinoma (OSCC).


International Journal of Molecular Sciences | 2015

Sinulariolide Suppresses Human Hepatocellular Carcinoma Cell Migration and Invasion by Inhibiting Matrix Metalloproteinase-2/-9 through MAPKs and PI3K/Akt Signaling Pathways

Yu-Jen Wu; Choo-Aun Neoh; Chia-Yu Tsao; Jui-Hsin Su; Hsing-Hui Li

Sinulariolide is an active compound isolated from the cultured soft coral Sinularia flexibilis. In this study, we investigate the migration and invasion effects of sinulariolide in hepatocellular carcinoma cell HA22T. Sinulariolide inhibited the migration and invasion effects of hepatocellular carcinoma cells in a concentration-dependent manner. The results of zymography assay showed that sinulariolide suppressed the activities of matrix metalloproteinase (MMP)-2 and MMP-9. Moreover, protein levels of MMP-2, MMP-9, and urokinase-type plasminogen activator (uPA) were reduced by sinulariolide in a concentration-dependent manner. Sinulariolide also exerted an inhibitory effect on phosphorylation of c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERK), phosphatidylinositol 3-kinase (PI3K), Akt, Focal adhesion kinase (FAK), growth factor receptor-bound protein 2 (GRB2). Taken together, these results demonstrated that sinulariolide could inhibit hepatocellular carcinoma cell migration and invasion and alter HA22T cell metastasis by reduction of MMP-2, MMP-9, and uPA expression through the suppression of MAPKs, PI3K/Akt, and the FAK/GRB2 signaling pathway. These findings suggest that sinulariolide merits further evaluation as a chemotherapeutic agent for human hepatocellular carcinoma.


Molecules | 2013

Sinulariolide Induced Hepatocellular Carcinoma Apoptosis through Activation of Mitochondrial-Related Apoptotic and PERK/eIF2α/ATF4/CHOP Pathway

Yi-Jen Chen; Jui-Hsin Su; Chia-Yu Tsao; Chun-Tzu Hung; Hsiang-Hao Chao; Jen-Jie Lin; Ming-Hui Liao; Zih-Yan Yang; Han Hisang Huang; Feng-Jen Tsai; Shun-Hsiang Weng; Yu-Jen Wu

Sinulariolide, an active compound isolated from the cultured soft coral Sinularia flexibilis, has potent anti-microbial and anti-tumorigenesis effects towards melanoma and bladder cancer cells. In this study, we investigated the effects of sinulariolide on hepatocellular carcinoma (HCC) cell growth and protein expression. Sinulariolide suppressed the proliferation and colony formation of HCC HA22T cells in a dose-dependent manner and induced both early and late apoptosis according to flow cytometry, Annexin V/PI stain and TUNEL/DAPI stain analyses. A mechanistic analysis demonstrated that sinulariolide-induced apoptosis was activated through a mitochondria-related pathway, showing up-regulation of Bax, Bad and AIF, and down- regulation of Bcl-2, Bcl-xL, MCl-1 and p-Bad. Sinulariolide treatment led to loss of the mitochondrial membrane potential, release of mitochondrial cytochrome c to the cytosol, and activation of both caspase-9 and caspase-3. Sinulariolide-induced apoptosis was significantly blocked by the caspase inhibitors Z-VAD-FMK and Z-DEVD-FMK. The increased expression of cleaved PARP also suggested that caspase-independent apoptotic pathway was involved. In the western blotting; the elevation of ER chaperones GRP78; GRP94; and CALR; as well as up-regulations of PERK/eIF2α/ATF4/CHOP; and diminished cell death with pre-treatment of eIF2α phosphatase inhibitor; salubrinal; implicated the involvement of ER stress-mediated PERK/eIF2α/ATF4/CHOP apoptotic pathway following sinulariolide treatment in hepatoma cells. The current study suggested sinulariolide-induced hepatoma cell cytotoxicity involved multiple apoptotic signal pathways. This may implicate that sinulariolide is a potential compound for the treatment of hepatocellular carcinoma.


Oral Oncology | 2013

Calreticulin, an endoplasmic reticulum-resident protein, is highly expressed and essential for cell proliferation and migration in oral squamous cell carcinoma

Wei-Fan Chiang; Tzer-Zen Hwang; Tzyh-Chyuan Hour; Lee-Hsin Wang; Chien-Chih Chiu; Hau-Ren Chen; Yu-Jen Wu; Chih-Chun Wang; Ling-Feng Wang; Chen-Yu Chien; Jen-Hao Chen; Chao-Tien Hsu; Jeff Yi-Fu Chen

OBJECTIVES Oral squamous cell carcinoma (OSCC) has emerged as one of the major malignant tumors of the head and neck cancers. However, the molecular mechanism behind tumorigenesis of OSCC is not fully understood. The aim of this study was to investigate the role of calreticulin (CRT), an endoplasmic reticulum-resident protein, in OSCC cells. MATERIALS AND METHODS Sixteen paired samples of tumor and non-cancerous matched tissue (NCMT), six OSCC cell lines and normal human oral keratinocytes (NHOKs), and oral tissue microarray were used to reveal the expression of CRT by Western blotting and immunohistochemistry. Later, shRNA-mediated stable knockdown of CRT in OSCC cells was generated. The knockdown cell line was used to analyze cell proliferation, colony formation, anchorage-independent growth and cell migration in vitro. RESULTS CRT was differentially expressed in fresh tumor samples and six OSCC cell lines but not adjacent NCMTs and NHOKs. In oral tissue microarray, we showed that there was positive CRT staining in the vast majority of tumor cases (99/103), in sharp contrast to that in NCMT cases (29/92) (p<0.001). Stable knockdown of CRT in oral cancer cells resulted in significantly reduced growth rate, colony-forming capacity and anchorage-independent growth. This may be attributed to the induction of G0/G1 cell cycle arrest when CRT was depleted in the cells. Both horizontal and vertical movements of the CRT-knockdown stable line were markedly impaired. The phosphorylation levels of focal adhesion kinase (FAK), paxillin and ERK1/2 and the activity of matrix metalloproteinase-2 and -9 (MMP-2 and MMP-9) were decreased in the CRT-knockdown cells. These results suggest that CRT can regulate oral cancer cell migration through activation of the FAK signaling pathway accompanied with proteolytic degradation of the extracellular matrix (ECM) by MMP-2 and MMP-9. CONCLUSION Together, this study has defined a novel biological role for CRT in oral cancer. CRT is a potential biomarker and may contribute to the malignant phenotypes of OSCC cells.


Marine Drugs | 2013

Proteomic Investigation of the Sinulariolide-Treated Melanoma Cells A375: Effects on the Cell Apoptosis through Mitochondrial-Related Pathway and Activation of Caspase Cascade

Hsing-Hui Li; Jui-Hsin Su; Chien-Chih Chiu; Jen-Jie Lin; Zih-Yan Yang; Wen-Ing Hwang; Yu-Kuei Chen; Yu-Hsuan Lo; Yu-Jen Wu

Sinulariolide is an active compound isolated from the cultured soft coral Sinularia flexibilis. In this study, we investigated the effects of sinulariolide on A375 melanoma cell growth and protein expression. Sinulariolide suppressed the proliferation and migration of melanoma cells in a concentration-dependent manner and was found to induce both early and late apoptosis by flow cytometric analysis. Comparative proteomic analysis was conducted to investigate the effects of sinulariolide at the molecular level by comparison between the protein profiles of melanoma cells treated with sinulariolide and those without treatment. Two-dimensional gel electrophoresis (2-DE) master maps of control and treated A375 cells were generated by analysis with PDQuest software. Comparison between these maps showed up- and downregulation of 21 proteins, seven of which were upregulated and 14 were downregulated. The proteomics studies described here identify some proteins that are involved in mitochondrial dysfunction and apoptosis-associated proteins, including heat shock protein 60, heat shock protein beta-1, ubiquinol cytochrome c reductase complex core protein 1, isocitrate dehydrogenase (NAD) subunit alpha (down-regulated), and prohibitin (up-regulated), in A375 melanoma cells exposed to sinulariolide. Sinulariolide-induced apoptosis is relevant to mitochondrial-mediated apoptosis via caspase-dependent pathways, elucidated by the loss of mitochondrial membrane potential, release of cytochrome c, and activation of Bax, Bad and caspase-3/-9, as well as suppression of p-Bad, Bcl-xL and Bcl-2. Taken together, our results show that sinulariolide-induced apoptosis might be related to activation of the caspase cascade and mitochondria dysfunction pathways. Our results suggest that sinulariolide merits further evaluation as a chemotherapeutic agent for human melanoma.


Marine Drugs | 2012

Induction of Apoptosis by Sinulariolide from Soft Coral through Mitochondrial-Related and p38MAPK Pathways on Human Bladder Carcinoma Cells

Choo-Aun Neoh; Robert Y.L. Wang; Zhong-Hao Din; Jui-Hsin Su; Yu-Kuei Chen; Feng-Jen Tsai; Shun-Hsiang Weng; Yu-Jen Wu

Sinulariolide, an isolated compound from the soft coral Sinularia flexibilis, possesses the anti-proliferative, anti-migratory and apoptosis-inducing activities against the TSGH bladder carcinoma cell. The anti-tumor effects of sinulariolide were determined by 3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, cell migration assay and flow cytometry, respectively. Sinulariolide inhibited the growth and migration of bladder carcinoma cells in a dose-dependent manner, as well as induced both early and late apoptosis as determined by the flow cytometer. Also, the sinulariolide-induced apoptosis is related to the mitochondrial-mediated apoptosis via caspase-dependent pathways, elucidated by the loss of mitochondrial membrane potential, release of cytochrome C, activation of caspase-3/-9, Bax and Bad, as well as suppression of Bcl-2/Bcl-xL/Mcl-1. Detection of the PARP-1 cleaved product suggested the partial involvement of caspase-independent pathways. Moreover, inhibition of p38MAPK activity leads to the rescue of the cell cytotoxicity of sinulariolide-treated TSGH cells, indicating that the p38MAPK pathway is also involved in the sinulariolide-induced cell apoptosis. Altogether, these results suggest that sinulariolide induces apoptosis against bladder cancer cells through mitochondrial-related and p38MAPK pathways.


Electrophoresis | 2012

Proteomic investigation of anti‐tumor activities exerted by sinularin against A2058 melanoma cells

Tzu-Rong Su; Jen-Jie Lin; Chien-Chih Chiu; Jeff Yi-Fu Chen; Jui-Hsin Su; Zhi-Jiao Cheng; Wen-Ing Hwang; Han Hsiang Huang; Yu-Jen Wu

The extracts from soft corals have been increasingly investigated for biomedical and therapeutic purposes. The aim of this study is to examine and analyze the anti‐tumor effects of the genus Sinularia extract sinularin on A2058 melanoma cells using MTT assay, cell migration assay, wound healing assay, flow cytometric analysis, and proteomic analysis. Sinularin dose‐dependently (1–5 μg/mL) inhibited melanoma cell proliferation while the treatment at identical concentrations suppressed cell migration. Sinularin dose‐dependently enhanced apoptotic melanoma cells and caused tumor cell accumulation at G2/M phase, indicating that sinularin exerts apoptosis‐induced and cell cycle‐delayed activities in A2058 melanoma cells. Comparative proteomic analysis was conducted to investigate the effects of sinularin at the molecular level by comparison between the protein profiling of melanoma cells treated with sinularin and without the treatment. Thirty‐five differential proteins (13 upregulated and 22 downregulated) concerning the treatment were identified by liquid chromatography‐tandem mass spectrometry. Proteomic data and Western blot displayed the levels of several tumor inhibitory or apoptosis‐associated proteins including annexin A1, voltage‐dependent anion‐selective channel protein 1 and prohibitin (upregulated), heat shock protein 60, heat shock protein beta‐1, and peroxiredoxin‐2 (downregulated) in A2058 melanoma cells exposed to sinularin. Increased expression of p53, cleaved‐caspase‐3, cleaved‐caspase‐8, cleaved‐caspase‐9, p21, and Bax and decreased expression of Bcl‐2 in sinularin‐treated melanoma cells suggest that the anti‐tumor activities of sinularin against melanoma cells are particularly correlated with these pro‐apoptotic factors. These data provide important information for the mechanisms of anti‐tumor effects of sinularin on melanoma cells and may be helpful for drug development and progression monitoring of human melanoma.


Marine Drugs | 2012

Induction of Apoptosis by 11-Dehydrosinulariolide via Mitochondrial Dysregulation and ER Stress Pathways in Human Melanoma Cells

Tzu-Rong Su; Feng-Jen Tsai; Jen-Jie Lin; Han Hsiang Huang; Chien-Chih Chiu; Jui-Hsin Su; Ya-Ting Yang; Jeff Yi-Fu Chen; Bing-Sang Wong; Yu-Jen Wu

In this study the isolated compound 11-dehydrosinulariolide from soft coral Sinularia leptoclados possessed anti-proliferative, anti-migratory and apoptosis-inducing activities against A2058 melanoma cells. Anti-tumor effects of 11-dehydrosinulariolide were determined by MTT assay, cell migration assay and flow cytometry. Growth and migration of melanoma cells were dose-dependently inhibited by 2–8 μg/mL 11-dehydrosinulariolide. Flow cytometric data indicated that 11-dehydrosinulariolide induces both early and late apoptosis in melanoma cells. It was found that the apoptosis induced by 11-dehydrosinulariolide is relevant to mitochondrial-mediated apoptosis via caspase-dependent pathways, elucidated by loss of mitochondrial membrane potential (∆Ψm), release of cytochrome C, activation of caspase-3/-9 and Bax as well as suppression of Bcl-2/Bcl-xL. The cleavage of PARP-1 suggested partial involvement of caspase-independent pathways. Immunoblotting data displayed up-regulations of PERK/eIF2α/ATF4/CHOP and ATF6/CHOP coupling with elevation of ER stress chaperones GRP78, GRP94, calnexin, calreticulin and PDI, implicating the involvement of these factors in ER stress-mediated apoptosis induced by 11-dehydrosinulariolide. The abolishment of apoptotic events after pre-treatment with salubrinal indicated that ER stress-mediated apoptosis is also induced by 11-dehydrosinulariolide against melanoma cells. The data in this study suggest that 11-dehydrosinulariolide potentially induces apoptosis against melanoma cells via mitochondrial dysregulation and ER stress pathways.


Journal of Proteomics | 2012

Proteomic profiling of the 11-dehydrosinulariolide-treated oral carcinoma cells Ca9-22: effects on the cell apoptosis through mitochondrial-related and ER stress pathway.

Chih-I Liu; Robert Yung-Liang Wang; Jen-Jie Lin; Jui-Hsin Su; Chien-Chih Chiu; Jiing-Chuan Chen; Jeff Yi-Fu Chen; Yu-Jen Wu

An oral squamous cell carcinoma Ca9-22 cell line was treated with 11-dehydrosinulariolide, an active compound isolated from the soft coral Sinularia leptoclados, in order to evaluate the effect of this compound on cell growth and protein expression. Cell proliferation was strongly inhibited by 11-dehydrosinulariolide treatment. The 2-DE master maps of control and treated Ca9-22 cells were generated by analysis with the PDQuest software. The comparison between such maps showed up- and down-regulation of 23 proteins, of which 14 were upregulated and 9 were downregulated. The proteomic studies described here have identified some proteins, which are involved in the mitochondrial dysfunction and ER-stress pathway and imply that 11-dehydrosinulariolide induces cell apoptosis through either mitochondrial dysfunction-related or ER stress pathway. Based on this observation, several proteins related to apoptosis pathway were explored for the potential roles involved in this drug-induced cytotoxicity. Furthermore, Salubrinal, an ER stress inhibitor, is able to protect the cell from 11-dehydrosinulariolide-induced apoptosis in a physiological dosage. The significance of these studies illustrates the potential development of anticancer drugs from the natural derivatives of soft coral.

Collaboration


Dive into the Yu-Jen Wu's collaboration.

Top Co-Authors

Avatar

Jeff Yi-Fu Chen

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Jen-Jie Lin

National Pingtung University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Chien-Chih Chiu

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tzyh-Chyuan Hour

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Tzu-Rong Su

Memorial Hospital of South Bend

View shared research outputs
Top Co-Authors

Avatar

Chen-Yu Chien

Kaohsiung Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge