King Chuen Wu
I-Shou University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by King Chuen Wu.
Phytomedicine | 2011
Jen Jyh Lin; Hui Ying Hsu; Jai Sing Yang; Kung Wen Lu; Rick Sai-Chuen Wu; King Chuen Wu; Tung Yuan Lai; Po Yuan Chen; Chia Yu Ma; W. Gibson Wood; Jing Gung Chung
We have shown that gypenosides (Gyp) induced cell cycle arrest and apoptosis in many human cancer cell lines. However, there are no reports showing that show Gyp acts on human leukemia HL-60 cells in vitro and in a murine xenograft model in vivo. In the present study effects of Gyp on cell morphological changes and viability, cell cycle arrest and induction of apoptosis in vitro and effects on Gyp in an in vivo murine xenograft model. Results indicated that Gyp induced morphological changes, decreased cell viability, induced G0/G1 arrest, DNA fragmentation and apoptosis (sub-G1 phase) in HL-60 cells. Gyp increased reactive oxygen species production and Ca(2+) levels but reduced mitochondrial membrane potential in a dose- and time-dependent manner. Gyp also changed one of the primary indicators of endoplasmic reticulum (ER) stress due to the promotion of ATF6-α and ATF4-α associated with Ca(2+) release. Gyp reduced the ratio of Bcl-2 to Bax due to an increase in the pro-apoptotic protein Bax and inhibited levels of the anti-apoptotic protein Bcl-2. Oral consumption of Gyp reduced tumor size of HL-60 cell xenograft mode mice in vivo. These results provide new information on understanding mechanisms by which Gyp induces cell cycle arrest and apoptosis in vitro and in vivo.
Integrative Cancer Therapies | 2012
Kung Wen Lu; Jung Chou Chen; Tung Yuan Lai; Jai Sing Yang; Shu Wen Weng; Yi Shih Ma; Hui-Yi Lin; Rick Sai Chuan Wu; King Chuen Wu; W. Gibson Wood; Jing Gung Chung
Purpose. Gypenosides (Gyp) are the major components of Gynostemma pentaphyllum Makino. The authors investigated the effects of Gyp on cell morphology, viability, cell cycle distribution, and induction of apoptosis in human oral cancer SAS cells and the determination of murine SAS xenograft model in vivo. Experimental design. Flow cytometry was used to quantify the percentage of viable cells; cell cycle distribution; sub-G1 phase (apoptosis); caspase-3, -8, and -9 activity; reactive oxygen species (ROS) production, intracellular Ca2+ determination; and the level of mitochondrial membrane potential (ΔΨm). Western blotting was used to examine levels of apoptosis-associated proteins, and confocal laser microscopy was used to examine the translocation of proteins in cells. Results. Gyp induced morphological changes, decreased the percentage of viable cells, caused G0/G1 phase arrest, and triggered apoptotic cell death in SAS cells. Cell cycle arrest induced by Gyp was associated with apoptosis. The production of ROS, increased intracellular Ca2+ levels, and the depolarization of ΔΨm were observed. Gyp increased levels of the proapoptotic protein Bax but inhibited the levels of the antiapoptotic proteins Bcl-2 and Bcl-xl. Gyp also stimulated the release of cytochrome c and Endo G. Translocation of GADD153 to the nucleus was stimulated by Gyp. Gyp in vivo attenuated the size and volume of solid tumors in a murine xenograft model of oral cancer. Conclusions. Gyp-induced cell death occurs through caspase-dependent and caspase-independent apoptotic signaling pathways, and the compound reduced tumor size in a xenograft nu/nu mouse model of oral cancer.
Science and Technology of Advanced Materials | 2013
King Chuen Wu; Ching Li Tseng; Chi Chang Wu; Feng Chen Kao; Yuan Kun Tu; Edmund Cheung So; Yang Kao Wang
Abstract Stem cells are known for their potential to repair damaged tissues. The adhesion, growth and differentiation of stem cells are likely controlled by the surrounding microenvironment which contains both chemical and physical cues. Physical cues in the microenvironment, for example, nanotopography, were shown to play important roles in stem cell fate decisions. Thus, controlling stem cell behavior by nanoscale topography has become an important issue in stem cell biology. Nanotechnology has emerged as a new exciting field and research from this field has greatly advanced. Nanotechnology allows the manipulation of sophisticated surfaces/scaffolds which can mimic the cellular environment for regulating cellular behaviors. Thus, we summarize recent studies on nanotechnology with applications to stem cell biology, including the regulation of stem cell adhesion, growth, differentiation, tracking and imaging. Understanding the interactions of nanomaterials with stem cells may provide the knowledge to apply to cell–scaffold combinations in tissue engineering and regenerative medicine.
Life Sciences | 2011
Edmund Cheung So; King Chuen Wu; Chia-Hua Liang; Jen Yin Chen; Sheng Nan Wu
AIMS PF573228 is an inhibitor of focal adhesion kinase (FAK) and recognized to affect cell adhesion and migration in many types of cells. Its effects on ion currents and membrane potential have been investigated in this study. MAIN METHOD Electrophysiological studies of PF573228 actions on ion currents were performed in pituitary tumor (GH(3)) cells, in GH(3) cells transfected with K(Ca)1.1 siRNAs and in human embryonic kidney (HEK) cells expressing α-human slowpoke (α-hSlo). KEY FINDINGS In whole-cell experiments, PF573228 reversibly increased the amplitude of Ca(2+)-activated K(+) currents (I(K(Ca))) in GH(3) cells. In inside-out recordings, this compound added to the bath did not modify single-channel conductance but stimulated large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels with an EC(50) value of 3.2 μM. As BK(Ca)-channel activity was stimulated by PF573228 (3 μM), subsequent application of BMS191011 (3 μM) did not further increase channel activity. PF573228 shifted the activation curve of BK(Ca) channels to less positive membrane potential. Change in the kinetic behavior of BK(Ca) channels caused by this compound is a result of the increased backward rate constants between closed states. PF573228 depressed the firing of action potentials in GH(3) cells. However, in GH(3) cells transfected with K(Ca)1.1 siRNAs, PF573228-stimulated I(K(Ca)) was abolished. In HEK293T cells expressing α-hSlo, PF573228 enhanced BK(Ca)-channel activity. SIGNIFICANCE In addition to an inhibition of FAK phosphorylation, PF573228 is effective in activating BK(Ca) channels. The direct stimulation of these channels by this compound may contribute to the underlying mechanism through which it influences cell behavior.
Anesthesia & Analgesia | 1996
Rick Sai-Chuen Wu; King Chuen Wu; Teresa Kit Man Wong; Ying-Huang Tsai; Robin Ka Shun Cheng; Peter P C Tan; Michael J. Bishop
This double-blind study investigates whether isoflurane/N2 O anesthesia adds to the bronchodilating effect of the beta2-adrenergic agonist, fenoterol, after an endotracheal tube (ETT)-induced increase in airway resistance. Forty-five patients with ASA physical status I-II were randomly assigned to two groups: fenoterol-treated patients (n = 23) were given three metered-dose inhaler puffs (600 micro gram) of fenoterol 10 min before induction of anesthesia and placebo-treated patients (n = 22) received three puffs of an aerosol containing no medication. Anesthesia was induced with thiopental and vecuronium intravenously. Respiratory system resistance (Rrs) was measured using a CP-100 pulmonary function monitor 5 min after endotracheal intubation. Inhalation anesthesia was then begun using 50% N2 O in O2 with end-tidal 1.3% isoflurane. Rrs measurements were repeated at 5, 15, and 30 min after the initiation of inhalation anesthesia. Postintubation Rrs was significantly lower in the fenoterol-treated patients than in the placebo-treated patients. Rrs declined by a mean of 17.1% after 30 min of inhalation anesthesia in the placebo-treated patients but declined by only 1.4% in the fenoterol-treated patients (P < 0.05 for fenoterol versus placebo). Our results confirm that fenoterol provides protection against ETT-induced increase of airway resistance. However, isoflurane, while a potent bronchodilator, does not add to the effect of fenoterol. (Anesth Analg 1996;83:238-41)
Nutrition and Cancer | 2011
Wen Tsong Hsieh; Hui-Yi Lin; Jou Hsuan Chen; Yueh-Hsiung Kuo; Ming Jen Fan; Rick Sai Chuan Wu; King Chuen Wu; W. Gibson Wood; Jing Gung Chung
Latex of Euphorbia antiquorum (EA) has inhibitory effects on several different cancer cell lines. However, the molecular mechanism of EA inhibitory effects on human cervical cancer HeLa cell growth has not been explored. EA induced apoptosis, which was characterized by morphological change, DNA fragmentation, increased sub-G1 population, and alterations in levels of apoptosis-associated proteins. Treatment with EA increased cell death and expression levels of caspase-8, -9, and -3. EA suppressed expression of Bcl-2, increased Bax, and reduced cleavage of Bid and the translocation of tBid to the mitochondria and the release of cytochrome c from mitochondria. EA caused a loss of mitochondrial membrane potential (ΔΨm) and an increase in cellular reactive oxygen species (ROS). EA-induced ROS formation was suppressed by cyclosporine A (an inhibitor of the ΔΨm) or allopurinol (an effective scavenger of ROS). EA also increased expression of Fas, FasL, and c-Jun N-terminal kinase (JNK), p38, and mitogen-activated protein kinase (MAPK) and decreased expression of extracellular signal-regulated kinase (ERK) 1/2-p. Co-treatment with the JNK inhibitor SP600125 inhibited EA-induced apoptosis and the activation of caspase-8, -9, and -3. Results of this study provide support for the hypothesis that EA causes cell death via apoptotic pathways in human cervical adenocarcinoma HeLa cells.
European Journal of Pharmacology | 2013
King Chuen Wu; Yi Hung Chen; Ka Shun Cheng; Yueh-Hsiung Kuo; Chin Tsang Yang; Kar Lok Wong; Yuan Kun Tu; Paul Chan; Yuk Man Leung
Imperatorin is a naturally occurring furocoumarin compound isolated from plants such as Angelica archangelica and Cnidium monnieri. It has multiple pharmacological effects including anticonvulsant effects. Here we determined the effects of imperatorin on voltage-gated Na(+) channels (VGSC) using whole-cell patch clamp techniques in differentiated neuronal NG108-15 cells. We showed that imperatorin inhibited VGSC; such inhibition did not show state-dependence. Imperatorin caused a left shift in the steady-state inactivation curve without affecting activation gating. The inhibition of VGSC by imperatorin displayed a mild frequency-dependence. Imperatorin was also shown to inhibit VGSC and action potential amplitude without affecting voltage-gated K(+) channels in rat hippocampal CA1 neurons. In conclusion, our results suggest that imperatorin dampens neuronal excitability by inhibiting VGSC.
Oncology Reports | 2016
Nou Ying Tang; Fu Shin Chueh; Chien Chih Yu; Ching Lung Liao; Jen Jyh Lin; Te Chun Hsia; King Chuen Wu; Hsin Chung Liu; Kung Wen Lu; Jing Gung Chung
Glioblastoma multiforme (GBM) is a highly malignant devastating brain tumor in adults. Benzyl isothiocyanate (BITC) is one of the isothiocyanates that have been shown to induce human cancer cell apoptosis and cell cycle arrest. Herein, the effect of BITC on cell viability and apoptotic cell death and the genetic levels of human brain glioblastoma GBM 8401 cells in vitro were investigated. We found that BITC induced cell morphological changes, decreased cell viability and the induction of cell apoptosis in GBM 8401 cells was time-dependent. cDNA microarray was used to examine the effects of BITC on GBM 8401 cells and we found that numerous genes associated with cell death and cell cycle regulation in GBM 8401 cells were altered after BITC treatment. The results show that expression of 317 genes was upregulated, and two genes were associated with DNA damage, the DNA-damage-inducible transcript 3 (DDIT3) was increased 3.66-fold and the growth arrest and DNA-damage-inducible α (GADD45A) was increased 2.34-fold. We also found that expression of 182 genes was downregulated and two genes were associated with receptor for cell responses to stimuli, the EGF containing fibulin-like extracellular matrix protein 1 (EFEMP1) was inhibited 2.01-fold and the TNF receptor-associated protein 1 (TRAP1) was inhibited 2.08-fold. BITC inhibited seven mitochondria ribosomal genes, the mitochondrial ribosomal protein; tumor protein D52 (MRPS28) was inhibited 2.06-fold, the mitochondria ribosomal protein S2 (MRPS2) decreased 2.07-fold, the mitochondria ribosomal protein L23 (MRPL23) decreased 2.08-fold, the mitochondria ribosomal protein S2 (MRPS2) decreased 2.07-fold, the mitochondria ribosomal protein S12 (MRPS12) decreased 2.08-fold, the mitochondria ribosomal protein L12 (MRPL12) decreased 2.25-fold and the mitochondria ribosomal protein S34 (MRPS34) was decreased 2.30-fold in GBM 8401 cells. These changes of gene expression can provide the effects of BITC on the genetic level and are potential biomarkers for glioblastoma therapy.
Environmental Toxicology | 2015
Rick Sai-Chuen Wu; King Chuen Wu; Chiu Chen Huang; Yi Ying Chiang; Chia Chen Chen; Ching Lung Liao; Chin Nan Chu; Jing Gung Chung
Various sedative agents, including dexmedetomidine (dex), induce immunosuppression, and enhance infection progression. However, there was no information on how anesthetic affects local and systemic cellular immune function. We conducted this study to examine the impact of dex on the differentiation and function of immune cells at site of inflammation and in peripheral blood during endotoxemia of mice. In BALB/c mice with and without endotoxemia, we evaluated the influence of two dosages of 5 and 50 mcg/kg/h intravenous dex on immune cells: including number of T cells (CD3), B cells (CD19), natural killer cells (CD8a), monocytes (CD11b), and macrophages (Mac‐3) in peripheral blood, the activities of macrophages in peripheral blood and in peritoneal lavage, and proliferation of B and T cells and of natural killer cells activity in the spleen. Endotoxemia increased the number of CD3 T cells, CD 19 B cells and macrophages in the peripheral blood, augmented macrophage activity in the peritoneum, and increased T cell proliferation and natural killer cell activity in the spleen. Further administration of 5 mcg/kg/h dex attenuated systemic increase in number of T cells, B cells, and macrophages during endotoxemia and 50 mcg/kg/h dex significantly attenuated the increase in activity of macrophages in the peripheral blood during endotoxemia. In the peritoneum, however, 5 mcg/kg/h dex preserved and 50 mcg/kg/h dexmedetomidine enhanced the activity of macrophages during endotoxemia. Increased in proliferation of T cells in spleen during endotoxemia was attenuated by both doses of dex. Last, 50 mcg/kg/h dex enhanced natural killer cells activity during endotoxemia. While preserving the effects of endotoxemia on macrophages activity in the infection site and natural killer cells activity in the spleen, dex decreased systemic fulminant immune reaction in endotoxemia, by attenuating the augmented response in the number of T cells, B cells and macrophages, activity of macrophages in the peripheral blood, and proliferation of T cells in spleen during endotoxemia.
European Journal of Pharmacology | 2014
Edmund Cheung So; King Chuen Wu; Feng Chen Kao; Sheng Nan Wu
Midazolam (MDL) was known to act through stimulation of benzodiazepine receptors (GABA). Whether midazolam affects ion currents and membrane potential in neurons remains largely unclear. Electrophysiological studies of midazolam actions were performed in differentiated motor neuron-like (NSC-34 and NG108-15) cells. Midazolam suppressed the amplitude of delayed rectifier K(+) current (IK(DR)) in a time- and concentration-dependent manner with an IC50 value of 10.4 µM. Addition of midazolam was noted to enhance the rate of IK(DR) inactivation. On the basis of minimal binding scheme, midazolam-induced block of IK(DR) was quantitatively provided with a dissociation constant of 9.8 µM. Recovery of IK(DR) from inactivation in the presence of midazolam was fitted by a single exponential. midazolam had no effect on M-type or erg-mediated K(+) current in these cells. Midazaolam (30 µM) suppressed the peak amplitude of voltage-gated Na(+) current (INa) with no change in the current-voltage relationships of this current. Inactivation kinetics of INa remained unaltered in the presence of this agent. In current-clamp configuration, midazolam (30 µM) prolonged the duration of action potentials (APs) and reduce AP amplitude. Similarly, in differentiated NG108-15 cells, the exposure to midazolam also suppressed IK(DR) with a concomitant increase in current inactivation. Midazolam can act as an open-channel blocker of delayed-rectifier K(+) channels in these cells. The synergistic blocking effects on IK(DR) and INa may contribute to the underlying mechanisms through which midazolam affects neuronal function in vivo.