Network


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

Hotspot


Dive into the research topics where Shu Jen Chang is active.

Publication


Featured researches published by Shu Jen Chang.


Mutation Research | 2012

Bufalin induces G0/G1 phase arrest through inhibiting the levels of cyclin D, cyclin E, CDK2 and CDK4, and triggers apoptosis via mitochondrial signaling pathway in T24 human bladder cancer cells

Wen Wen Huang; Jai Sing Yang; Shu Jen Pai; Ping Ping Wu; Shu Jen Chang; Fu Shin Chueh; Ming Jen Fan; Shang Ming Chiou; Hsiu Maan Kuo; Chin Chung Yeh; Po Yuan Chen; Minoru Tsuzuki; Jing Gung Chung

Most of the chemotherapy treatments for bladder cancer aim to kill the cancer cells, but a high recurrence rate after medical treatments is still occurred. Bufalin from the skin and parotid venom glands of toad has been shown to induce apoptotic cell death in many types of cancer cell lines. However, there is no report addressing that bufalin induced cell death in human bladder cancer cells. The purpose of this study was investigated the mechanisms of bufalin-induced apoptosis in a human bladder cancer cell line (T24). We demonstrated the effects of bufalin on the cell growth and apoptosis in T24 cells by using DAPI/TUNEL double staining, a PI exclusion and flow cytometric analysis. The effects of bufalin on the production of reactive oxygen species (ROS), the level of mitochondrial membrane potential (ΔΨ(m)), and DNA content including sub-G1 (apoptosis) in T24 cells were also determined by flow cytometry. Western blot analysis was used to examine the expression of G(0)/G(1) phase-regulated and apoptosis-associated protein levels in bufalin-treated T24 cells. The results indicated that bufalin significantly decreased the percentage of viability, induced the G(0)/G(1) phase arrest and triggered apoptosis in T24 cells. The down-regulation of the protein levels for cyclin D, CDK4, cyclin E, CDK2, phospho-Rb, phospho-AKT and Bcl-2 with the simultaneous up-regulation of the cytochrome c, Apaf-1, AIF, caspase-3, -7 and -9 and Bax protein expressions and caspase activities were observed in T24 cells after bufalin treatment. Based on our results, bufalin induces apoptotic cell death in T24 cells through suppressing AKT activity and anti-apoptotic Bcl-2 protein as well as inducing pro-apoptotic Bax protein. The levels of caspase-3, -7 and -9 are also mediated apoptosis in bufalin-treated T24 cells. Therefore, bufalin might be used as a therapeutic agent for the treatment of human bladder cancer in the future.


Neurochemical Research | 2009

Baicalein-Induced Apoptosis via Endoplasmic Reticulum Stress Through Elevations of Reactive Oxygen Species and Mitochondria Dependent Pathway in Mouse-Rat Hybrid Retina Ganglion Cells (N18)

Yu Ching Li; Hui Ju Lin; Jen Hung Yang; Jai Sing Yang; Heng Chien Ho; Shu Jen Chang; Te Chun Hsai; Hsu Feng Lu; An Cheng Huang; Jing Gung Chung

Studies were designed to investigate the effects of baicalein on mouse–rat hybrid retina ganglion cells (N18) to better understand its effect on apoptosis and apoptosis-related genes in vitro. Cell viability, reactive oxygen species (ROS), cytoplasmic Ca2+, mitochondrial membrane potential (MMP), apoptosis induction, and caspases-3 activity were examined by flow cytometric assay. Apoptosis-associated proteins such as p53, Bax, Bcl-2, cytochrome c, and caspase-3 were examined by Western blot. We demonstrated the increase in the levels of p53, Bax, and cytochrome c and decrease in the level of Bcl-2, which are associated with the induction of apoptotic cell death after 24 h treatment with baicalein in N18 cells. Baicalein induced an increase in the cytoplasmic levels of ROS and Ca2+ in 1 h and reached their peak at 3 h, and thereafter a loss of MMP by flow cytometry. We also demonstrated a release of the cytochrome c from mitochondria into cytosol and an activation of caspase-3, which led to the occurrence of apoptosis in N18 cells treated with baicalein by Western blot. Pretreatment was conducted with BAPTA (intracellular calcium chelator) in baicalein-treated cells, the decline of MMP was recovered, and the increase in the level of cytoplasmic Ca2+ was suppressed, and the proportion of apoptosis was also markedly diminished. In conclusion, our data suggests that oxidative stress and cellular Ca2+ modulates the baicalein-induced cell death via a Ca2+-dependent mitochondrial death pathway in N18 cells.


International Journal of Oncology | 2013

Tetrandrine induces cell death in SAS human oral cancer cells through caspase activation-dependent apoptosis and LC3-I and LC3-II activation-dependent autophagy

An Cheng Huang; Jin-Cherng Lien; Meng Wei Lin; Jai Sing Yang; Ping Ping Wu; Shu Jen Chang; Tung Yuan Lai

Numerous studies have demonstrated that autophagy is associated with cancer development. Thus, agents to induce autophagy could be employed in some cases for the treatment of cancer. Our results showed that tetrandrine significantly decreased the viability of SAS cells in a concentration- and time-dependent manner. Tetrandrine induced nuclear condensation, demonstrated by DAPI staining. The early events in apoptosis analysed by Annexin V/PI staining indicated that the percentage of cells staining positive for Annexin V was slightly increased in SAS cells with tetrandrine treatment but was much lower following bafilomycin A1 pre-treatment. Tetrandrine caused AVO and MDC induction in SAS cells in a concentration-dependent manner by fluorescence microscopy. Tetrandrine also caused LC-3 expression in SAS cells in a time-dependent manner. Our results show that tetrandrine treatment induced the levels of cleaved caspase-3 in a concentration- and time-dependent manner. Tetrandrine treatment induced the levels of LC-3 II, Atg-5, beclin-1, p-S6, p-ULK, p-mTOR, p-Akt (S473) and raptor. Tetrandrine decreased cell viability, but bafilomycin A1, 3-MA, chloroquine and NAC protected tetrandrine-treated SAS cells against decrease of cell viability. Atg-5, beclin-1 siRNA decreased tetrandrine-induced cleaved caspase-3 and cleaved PARP in SAS cells and protected tetrandrine-treated SAS cells against decrease in cell viability. Chloroquine, NAC and bafilomycin A1 also decreased tetrandrine-induced cleaved caspase-3 and cleaved PARP in SAS cells. Our results indicate the tetrandrine induces apoptosis and autophagy of SAS human cancer cells via caspase-dependent and LC3-I and LC3-II‑dependent pathways.


Environmental Toxicology | 2015

Bufalin inhibits migration and invasion in human hepatocellular carcinoma SK‐Hep1 cells through the inhibitions of NF‐kB and matrix metalloproteinase‐2/‐9‐signaling pathways

Ya Yin Chen; Hsu Feng Lu; Shu Chun Hsu; Chao Lin Kuo; Shu Jen Chang; Jen Jyh Lin; Ping Ping Wu; Jia You Liu; Ching Hsiao Lee; Jing Gung Chung; Jin Biou Chang

Metastasis plays an important role in mortality of cancer patients. Migration and invasion are the major characteristics of tumor metastasis. The induction of matrix metalloproteinases (MMPs) such as MMP‐2 and ‐9 are particularly important for the invasiveness of various cancer cells. Bufalin, a class of toxic steroids, was purified from the skin glands of Bufo gargarizans or Bufo melanostictus; it is known to inhibit proliferation of human cancer cells. In this study, we investigated the antiinvasive mechanisms of bufalin in the human hepatocellular cancer cell line SK‐Hep1. Bufalin significantly reduced serum‐induced cell invasion and migration. Furthermore, bufalin markedly inhibited MMP‐2 and ‐9 activity, mRNA expression and protein levels in SK‐Hep1 cells. Bufalin attenuated phosphoinisitide‐3‐kinase (PI3K) and phosphorylation of AKT which was associated with reduced levels of nuclear factor kappa B (NF‐κB). Bufalin also suppressed protein levels of FAK and Rho A, VEGF, MEKK3, MKK7, and uPA and it diminished NF‐κB translocation. Based on these observations, we propose that bufalin is acts as an antiinvasive agent by inhibiting MMP‐2 and ‐9 and involving PI3K/AKT and NF‐κB pathways. Bufalin is a potential therapeutic agent that may have efficacy in preventing the invasion and metastasis of malignant liver tumors.


Evidence-based Complementary and Alternative Medicine | 2013

Suppressions of Migration and Invasion by Cantharidin in TSGH-8301 Human Bladder Carcinoma Cells through the Inhibitions of Matrix Metalloproteinase-2/-9 Signaling

Yi Ping Huang; Chien Hang Ni; Chi Cheng Lu; Jo Hua Chiang; Jai Sing Yang; Yang Ching Ko; Jing Pin Lin; Jehn Hwa Kuo; Shu Jen Chang; Jing Gung Chung

Cancer metastasis becomes an initial cause of cancer death in human population. In many cancers, it has been shown that the high levels of matrix metalloproteinase (MMP)-2 and/or MMP-9 are associated with the invasive phenotypes of cancer cells. In this study, we investigated the effects of cantharidin, a derivative of blister beetles which is one of the traditional Chinese medicines, on the adhesion, migration, and invasion of human bladder cancer TSGH-8301 cells. Cantharidin effectively suppressed TSGH-8301 cell adhesion, migration, and invasion in a concentration-dependent manner. Results from Western blotting, RT-PCR, and gelatin zymography assays indicated that cantharidin blocked the protein levels, gene expression (mRNA), and activities of MMP-2 and -9 in TSGH-8301 cells. Cantharidin also significantly suppressed the protein expressions of p-p38 and p-JNK1/2 in TSGH-8301 cells. Taken together, cantharidin was suggested to present antimetastatic potential via suppressing the levels of MMP-2 and MMP-9 expression that might be mediated by targeting the p38 and JNK1/2 MAPKs pathway in TSGH-8301 human bladder cancer cells.


Oncology Reports | 2012

Anti-inflammatory effects of Calophyllum inophyllum L. in RAW264.7 cells

Shih Chang Tsai; Yu Hsin Liang; Jo Hua Chiang; Fon Chang Liu; Wen Hsin Lin; Shu Jen Chang; Wei Yu Lin; Chieh Hsi Wu; Jing Ru Weng

Calophyllum inophyllum L. has been used as folk medicine in the treatment of ocular burn and it has demonstrated potential to be an anti-inflammatory agent. The aim of this study was to explore the anti-inflammatory activities of an acetone extract of C. inophyllum L. leaves (CIL). The CIL extract was tested on lipopolysaccharide (LPS)-induced RAW 264.7 cells to evaluate the effect of CIL extract on the expression of nitric oxide (NO) and inducible nitric oxide synthase (iNOS). Results showed that the CIL extract markedly suppressed the LPS-induced production of nitric oxide, as well as the expression of iNOS, cyclooxygenase (COX)-2 and nuclear factor-kappaB (NF-κB) in a dose-dependent manner. LPS-induced microRNA (miR)-146a expression was inhibited by CIL extract, while miR-155 and miR-424 expression was not affected as demonstrated using quantitative RT-PCR analysis. Taken together, these observations show that CIL extract has anti-inflammatory effect, which extends the potential application for prevention of inflammatory diseases, and its mechanism may be partially associated with blocking COX-2 and iNOS of RAW 264.7 cells.


Environmental Toxicology | 2017

Tetrandrine induces programmed cell death in human oral cancer CAL 27 cells through the reactive oxygen species production and caspase-dependent pathways and associated with beclin-1-induced cell autophagy

Jin-Cherng Lien; Meng Wei Lin; Shu Jen Chang; Kuang Chi Lai; An Cheng Huang; Fu Shun Yu; Jing Gung Chung

Tetrandrine, a bisbenzylisoquinoline alkaloid, is extracted from the root of the Chinese herb Radix Stephania tetrandra S Moore. This compound has antitumor activity in different cancer cell types. In this study, the effects of tetrandrine on human oral cancer CAL 27 cells were examined. Results indicated that tetrandrine induced cytotoxic activity in CAL 27 cells. Effects were due to cell death by the induction of apoptosis and accompany with autophagy and these effects were concentration‐ and time‐dependent manners. Tetrandrine induced apoptosis was accompanied by alterations in cell morphology, chromatin fragmentation, and caspase activation in CAL 27 cells. Tetrandrine treatment also induced intracellular accumulation of reactive oxygen species (ROS). The generation of ROS may play an important role in tetrandrine‐induced apoptosis. Tetrandrine triggered LC3B expression and induced autophagy in CAL 27 cells. Tetrandrine induced apoptosis and autophagy were significantly attenuated by N‐acetylcysteine pretreatment that supports the involvement of ROS production. Tetrandrine induced cell death may act through caspase‐dependent apoptosis with Beclin‐1‐induced autophagy in human oral cancer cells.


Environmental Toxicology | 2016

Tetrandrine induces apoptosis Via caspase-8, -9, and -3 and poly (ADP ribose) polymerase dependent pathways and autophagy through beclin-1/ LC3-I, II signaling pathways in human oral cancer HSC-3 cells

Fu Shun Yu; Chun Shu Yu; Jaw Chyun Chen; Jiun Long Yang; Hsu Feng Lu; Shu Jen Chang; Meng Wei Lin; Jing Gung Chung

Tetrandrine is a bisbenzylisoquinoline alkaloid that was found in the Radix Stephania tetrandra S Moore. It had been reported to induce cytotoxic effects on many human cancer cells. In this study, we investigated the cytotoxic effects of tetrandrine on human oral cancer HSC‐3 cells in vitro. Treatments of HSC‐3 cells with tetrandrine significantly decreased the percentage of viable cells through the induction of autophagy and apoptosis and these effects are in concentration‐dependent manner. To define the mechanism underlying the cytotoxic effects of tetrandrine, we investigated the critical molecular events known to regulate the apoptotic and autophagic machinery. Tetrandrine induced chromatin condensation, internucleosomal DNA fragmentation, activation of caspases‐3, ‐8, and ‐9, and cleavage of poly (ADP ribose) polymerase (PARP) that were associated with apoptosis, and it also enhanced the expression of LC3‐I and ‐II that were associated with the induction of autophagy in human squamous carcinoma cell line (HSC‐3) cells. Tetrandrine induced autophagy in HSC‐3 cells was significantly attenuated by bafilomycin A1 (inhibitor of autophagy) pre‐treatment that confirmed tetrandrine induced cell death may be associated with the autophagy. In conclusion, we suggest that tetrandrine induced cell death may be through the induction of apoptosis as well as autophagy in human oral cancer HSC‐3 cells via PARP, caspases/Becline I/LC3‐I/II signaling pathways.


Phytomedicine | 2013

Elucidating the inhibitory mechanisms of the ethanolic extract of the fruiting body of the mushroom Antrodia cinnamomea on the proliferation and migration of murine leukemia WEHI-3 cells and their tumorigenicity in a BALB/c allograft tumor model

Fon Chang Liu; Ming Tsung Lai; Ying Yi Chen; Wen-Hsin Lin; Shu Jen Chang; Ming Jyh Sheu; Chieh Hsi Wu

The aim of this study was to explore whether the ethanolic extract of Antrodia cinnamomea (EEAC), a medical mushroom form Taiwan, could affect the proliferation and migration of WEHI-3 cells in vitro and to explore the antitumor effects of EEAC in BALB/c mice engrafted with WEHI-3 cells. The results showed that EEAC inhibited the proliferation of WEHI-3 cells, resulting in the accumulation of cell in G0/G1 and G2/M phases, as determined by flow cytometry. Moreover, EEAC markedly reduced the migration of WEHI-3 cells, as determined by a transwell assay. Treatment of WEHI-3 cells with EEAC also decreased MMP-9 protein expression and enzyme activity. The protein levels of p-Akt, p-ERK1/2 were also decreased, whereas the expression of p21 and p27 was increased. Furthermore, in an in vivo model, EEAC treatment reduced the infiltration of WEHI-3 cells into the liver and spleens and decreased tumor growth. Other bioactive compounds, such as cordycepin and zhankuic acid A, have been demonstrated to reduce the expression of MMP-9, cyclin E, cyclin D1 and to increase the expression of p21, p27. This is the first study to investigate that the mechanisms by which EEAC reduce the proliferation and migration of WEHI-3 cells in vitro, as well as the ability of EEAC to reduced infiltration of WEHI-3 cells into the liver and spleen in vivo. The results suggest that EEAC may prove to be useful in future antileukemic therapies.


Environmental Toxicology | 2017

Phenethyl isothiocyanate alters the gene expression and the levels of protein associated with cell cycle regulation in human glioblastoma GBM 8401 cells

Yu Cheng Chou; Meng Ya Chang; Mei Jen Wang; Hsin Chung Liu; Shu Jen Chang; Tomor Harnod; Chih Huang Hung; Hsu Tung Lee; Chiung Chyi Shen; Jing Gung Chung

Glioblastoma is the most common and aggressive primary brain malignancy. Phenethyl isothiocyanate (PEITC), a member of the isothiocyanate family, can induce apoptosis in many human cancer cells. Our previous study disclosed that PEITC induces apoptosis through the extrinsic pathway, dysfunction of mitochondria, reactive oxygen species (ROS)‐induced endoplasmic reticulum (ER) stress, and intrinsic (mitochondrial) pathway in human brain glioblastoma multiforme (GBM) 8401 cells. To the best of our knowledge, we first investigated the effects of PEITC on the genetic levels of GBM 8401 cells in vitro. PEITC may induce G0/G1 cell‐cycle arrest through affecting the proteins such as cdk2, cyclin E, and p21 in GBM 8401 cells. Many genes associated with cell‐cycle regulation of GBM 8401 cells were changed after PEITC treatment: 48 genes were upregulated and 118 were downregulated. The cell‐division cycle protein 20 (CDC20), Budding uninhibited by benzimidazole 1 homolog beta (BUB1B), and cyclin B1 were downregulated, and clusterin was upregulated in GBM 8401 cells treated with PEITC. These changes of gene expression can provide the effects of PEITC on the genetic levels and potential biomarkers for glioblastoma.

Collaboration


Dive into the Shu Jen Chang's collaboration.

Top Co-Authors

Avatar

Hsu Feng Lu

Fu Jen Catholic University

View shared research outputs
Top Co-Authors

Avatar

Jo Hua Chiang

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar

Chi Cheng Lu

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

Jin Biou Chang

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ming Tsung Lai

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Ya Yin Chen

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Bin Chuan Ji

Chienkuo Technology University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chuan Sung Chiu

Hsin Sheng College of Medical Care and Management

View shared research outputs
Researchain Logo
Decentralizing Knowledge