Network


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

Hotspot


Dive into the research topics where Shu-Jem Su is active.

Publication


Featured researches published by Shu-Jem Su.


Toxicology and Applied Pharmacology | 2003

Cardiotoxin-III selectively enhances activation-induced apoptosis of human CD8+ T lymphocytes

Shu-Hui Su; Shu-Jem Su; Shinne-Ren Lin; Kee-Lung Chang

Cardiotoxin-III (CTX-III), a major cardiotoxin isolated from the venom of the Taiwan cobra (Naja naja atra), is a highly basic, hydrophobic, toxic protein, which can induce lysis of mononuclear cells by an unknown mechanism. This study was undertaken to investigate the effects of CTX-III on untreated and PHA-activated peripheral blood mononuclear cells (PBMCs) in vitro. The results show that treatment of PHA-activated lymphocytes with CTX-III (10 microg/ml) induced apoptosis and depletion of the CD8(+) population. In both untreated and PHA-treated lymphocytes, interferon-gamma production was dramatically reduced and interleukin-2 (IL-2) production was moderately reduced by CTX-III treatment. In PHA-activated lymphocytes, CD4 expression was increased, whereas CD8 and IL-2R beta chain (CD25) expression were decreased. In contrast, CTX-III had no effect on the viability of PHA-activated monocytes but significantly enhanced their tumor necrosis factor-alpha production. These results show that CTX-III selectively enhanced activation-induced apoptosis in CD8(+) T cells. CTX-III was found to bind to the cell membrane of PHA-stimulated PBMCs, and three CTX-III-binding proteins, with molecular weights of 92, 77, and 68 kDa, were identified. We therefore propose that CTX-III interacts with one or more cell surface proteins and initiates a signal pathway causing functional changes. These findings provide an insight into the immunomodulatory properties of CTX-III and suggest a novel method for the selective induction of apoptosis in CD8(+) T lymphocytes.


Cancer Letters | 2009

Combined effects of terazosin and genistein on a metastatic, hormone-independent human prostate cancer cell line

Kee-Lung Chang; Hsiao-Ling Cheng; Li-Wen Huang; Bau-Shan Hsieh; Yu-Chen Hu; Tsai-Tung Chih; Huey-Wen Shyu; Shu-Jem Su

Metastatic prostate cancer progresses from androgen-dependent to androgen-independent. Terazosin, a long-acting selective alpha1-adrenoreceptor antagonist, induces apoptosis of prostate cancer cells in an alpha1-adrenoreceptor-independent manner, while genistein, a major soy isoflavone, inhibits the growth of several types of cancer cells. The present study was designed to test the therapeutic potential of a combination of terazosin and genistein using a metastatic, hormone-independent prostatic cancer cell line, DU-145. Terazosin or genistein treatment inhibited the growth of DU-145 cells in a dose-dependent manner, whereas had no effect on normal prostate epithelial cells. Addition of 1 microg/ml of terazosin, which was inactive alone, augmented the growth inhibitory effect of 5 microg/ml of genistein. Co-treatment with terazosin resulted in the genistein-induced arrest of DU-145 cells in G2/M phase being overridden and an increase in apoptotic cells, as evidenced by procaspase-3 activation and PARP cleavage. The combination also caused a greater decrease in the levels of the apoptosis-regulating protein, Bcl-XL, and of VEGF165 and VEGF121 than genistein alone. In conclusion, the terazosin/genistein combination was more effective in inhibiting cell growth and VEGF expression as well as inducing apoptosis of the metastatic, androgen-independent prostate cancer cell line, DU-145, than either alone. The doses used in this study are in lower and nontoxic anticancer dosage range, suggesting this combination has potential for therapeutic use.


Molecular Cancer | 2010

Arecoline induces HA22T/VGH hepatoma cells to undergo anoikis - involvement of STAT3 and RhoA activation

Hsiao-Ling Cheng; Shu-Jem Su; Li-Wen Huang; Bau-Shan Hsieh; Yu-Chen Hu; Thu-Ching Hung; Kee-Lung Chang

BackgroundOur previous study showed that, in basal cell carcinoma cells, arecoline reduces levels of the tumor cell survival factor interleukin-6 (IL-6), increases levels of tumor suppressor factor p53, and elicits cell cycle arrest, followed by apoptosis. In preliminarily studies, we observed that arecoline induces detachment of the human-derived hepatoma cell line HA22T/VGH from the extracellular matrix. In the present study, we explored the fate of the detached HA22T/VGH cells and investigated the underlying mechanism.MethodsHA22T/VGH cells or primary cultured rat hepatocytes were treated with arecoline, then changes in morphology, viability, apoptosis, and the expression of surface β1-integrin, apoptosis-related proteins, and IL-6 were examined. Furthermore, activation of the signal transducer and activator of transcription 3 (STAT3) pathway and the RhoA/Rock signaling pathway, including p190RhoGAP and Src homology-2 domain-containing phosphatase SHP2, was examined.ResultsA low concentration of arecoline (≤ 100 μg/ml) caused cytoskeletal changes in HA22T/VGH cells, but not hepatocytes, and this was accompanied by decreased β1-integrin expression and followed by apoptosis, indicating that HA22T/VGH cells undergo anoikis after arecoline treatment. IL-6 expression and phosphorylation of STAT3, which provides protection against anoikis, were inhibited and levels of downstream signaling proteins, including Bcl-XL and Bcl-2, were decreased, while Bax expression, mitochondrial cytochrome c release, and caspase-3 activity were increased. In addition, phosphorylation/activation of p190RhoGAP, a RhoA inhibitor, and of its upstream regulator, SHP2, was inhibited by arecoline treatment, while Rho/Rock activation was increased. Addition of the RhoA inhibitor attenuated the effects of arecoline.ConclusionsThis study demonstrated that arecoline induces anoikis of HA22T/VGH cells involving inhibition of STAT3 and increased RhoA/Rock activation and that the STAT3 and RhoA/Rock signaling pathways are connected.


Free Radical Biology and Medicine | 2014

Phenethyl isothiocyanate induces DNA damage-associated G2/M arrest and subsequent apoptosis in oral cancer cells with varying p53 mutations.

Yao-Tsung Yeh; Hua Yeh; Shu-Hui Su; Jian-Sheng Lin; Kuo-Jui Lee; Huey-Wen Shyu; Zi-Feng Chen; Sheng-Yun Huang; Shu-Jem Su

Phenethyl isothiocyanate (PEITC) is a naturally occurring cruciferous vegetable-derived compound that inhibits cell growth and induces apoptosis in oral cancer cells. However, the exact mechanism of PEITC action has not been fully elucidated. This study investigated the molecular mechanism and anticancer potential of PEITC in oral squamous cell carcinoma (OSCC) cells with various p53 statuses. PEITC inhibited the growth of OC2, SCC4, and SCC25 cells (functional p53 mutants) in a dose-dependent manner with low toxicity to normal cells. Treatment with PEITC induced reactive oxygen species production, nitric oxide generation, and GSH depletion and triggered DNA damage response as evidenced by flow cytometry, 8-OHdG formation, and comet assay. Furthermore, the subsequent activation of ATM, Chk2, and p53 as well as the increased expression of downstream proteins p21 and Bax resulted in a G2/M phase arrest by inhibiting Cdc25C, Cdc2, and cyclin B1. The PEITC-induced apoptotic cell death, following a diminished mitochondrial transmembrane potential, reduced the expression of Bcl-2 and Mcl-1, released mitochondrial cytochrome c, and activated caspase 3 and PARP cleavage. The p53 inhibitor pifithrin-α and the antioxidants N-acetylcysteine and glutathione (GSH) protected the cells from PEITC-mediated apoptosis. However, mito-TEMPO, catalase, apocynin, and L-NAME did not prevent PEITC-induced cell death, suggesting that PEITC induced G2/M phase arrest and apoptosis in oral cancer cells via a GSH redox stress and oxidative DNA damage-induced ATM-Chk2-p53-related pathway. These results provide new insights into the critical roles of both GSH redox stress and p53 in the regulation of PEITC-induced G2/M cell cycle arrest and apoptosis in OSCCs.


Nutrition | 2013

Combined effect of soy isoflavones and vitamin D3 on bone loss in ovariectomized rats

Kee-Lung Chang; Yu-Chen Hu; Bau-Shan Hsieh; Hsiao-Ling Cheng; Hen-Wei Hsu; Li-Wen Huang; Shu-Jem Su

OBJECTIVE Several studies have shown that soy isoflavones have estrogen-like activities and might constitute an alternative to hormone replacement treatment. The present study investigated the effects of soy isoflavones alone and combined with vitamin D3 on prevention of bone loss. METHODS Sprague-Dawley rats were sham-operated (n = 8) or ovariectomized (OVX; n = 40), and then the OVX rats were randomly assigned to five groups that were untreated or treated for 14 wk with vitamin D3, 17β-estradiol, soy isoflavone extract (SIE), or vitamin D3 plus SIE. The effects of the isoflavones and 1α,25(OH)(2)D(3) on cultured osteoblasts and osteoclasts also were investigated. RESULTS In OVX rats, the bone mineral density and trabecular bone volume loss were improved by 17β-estradiol, SIE, or SIE plus vitamin D3 treatment. SIE treatment was more effective than vitamin D3 or 17β-estradiol in inhibiting increases in serum tumor necrosis factor-α levels and osteoblast osteoprotegerin expression. SIE plus vitamin D3 was more effective in increasing osterix expression than each alone. Bone cell cultures showed that the isoflavones induced preosteoblasts to differentiate into osteoblasts and increased osteoblast mineralization. Isoflavones inhibited preosteoclasts and osteoclast proliferation and decreased osteoclast resorption. The combination of isoflavones plus 1α,25(OH)(2)D(3) showed additive effects on the increase in cell proliferation of cultured preosteoblasts. CONCLUSION Treatment with soy isoflavones might be an alternative to hormone replacement therapy in decreasing bone loss from postmenopausal estrogen deficiency. In addition, there are further effects on increasing transcription factor osterix expression and preosteoblast proliferation when these were combined with vitamin D3.


International Journal of Cardiology | 2011

Hemeoxygenase-1 expression in response to arecoline-induced oxidative stress in human umbilical vein endothelial cells

Thu-Ching Hung; Li-Wen Huang; Shu-Jem Su; Bau-Shan Hsieh; Hsiao-Ling Cheng; Yu-Chen Hu; Yen-Hui Chen; Chi-Ching Hwang; Kee-Lung Chang

BACKGROUND Arecoline, the most abundant areca alkaloid, has been reported to stimulate reactive oxygen species (ROS) production in several cell types. Overproduction of ROS has been implicated in atherogenesis. Hemeoxygenase-1 (HO-1) has cytoprotective activities in vascular tissues. This study investigated the effect of arecoline on adhesion molecule expression and explored the role of HO-1 in this process. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with arecoline, then ROS levels and the expression of adhesion molecules and HO-1 were analyzed and potential signaling pathways investigated. RESULTS After 2h of arecoline treatment, ROS production was stimulated and reached a maximum at 12h. Expression of the adhesion molecules ICAM and VCAM was also induced. Glutathione pretreatment completely blocked arecoline-stimulated ROS production and VCAM expression, but not ICAM expression. Arecoline also induced HO-1 expression and this effect was partly due by ROS stimulation. Inhibition of c-jun N-terminal kinase (JNK) by SP600125, p38 by SB 203580, or tyrosine kinase by genistein reduced arecoline-induced HO-1 expression. In contrast, inhibition of ERK (extracellular signal-related MAP kinase) by PD98059 had no effect. Transfection of HUVECs with the GFP/HO-1 gene, which resulted in a 5-fold increase in HO-1 activity, markedly, but not completely, inhibited the decrease in cell viability caused by arecoline. CONCLUSIONS This study demonstrates that, in HUVECs, arecoline stimulates ROS production and ICAM and VCAM expression. HO-1 expression is also upregulated through the ROS, tyrosine kinase, and MAPK (JNK and p38) signaling pathways.


Journal of Nutritional Biochemistry | 2011

Combined arginine and ascorbic acid treatment induces apoptosis in the hepatoma cell line HA22T/VGH and changes in redox status involving the pentose phosphate pathway and reactive oxygen and nitrogen species

Bau-Shan Hsieh; Li-Wen Huang; Shu-Jem Su; Hsiao-Ling Cheng; Yu-Chen Hu; Thu-Ching Hung; Kee-Lung Chang

Arginine is a physiological substrate for nitric oxide synthase to generate nitric oxide (NO), which can influence tumor cell survival, while ascorbic acid is selectively toxic for cancer cells. This study explored the effect of an arginine/ascorbic acid combination on human cancer cell lines. The hepatoma cell line HA22T/VGH was the most sensitive of the tested cells to combination treatment. A combination of 5.74 mM of arginine and 0.57 mM of ascorbic acid induced HA22T/VGH cell death through apoptosis and an increase in levels of reactive oxygen species and NO, as well as its stable products NO(2)(-) and NO(3)(-). The combination also reduced the activity of glucose-6-phosphate dehydrogenase, 6-phosphogluconate dehydrogenase, and transaldolase in the pentose phosphate pathway, a major mechanism for producing NADPH, resulting in a marked decrease in intracellular NADPH levels. A dramatic decrease in intracellular glutathione (GSH) levels, a decrease in the mitochondrial membrane potential, ATP depletion and release of cytochrome c were also seen. Caspase-9 and caspase-3 were activated, apoptotic protein Bax expression increased and the expression of the anti-apoptotic proteins Bcl-2 and Bcl-xL decreased. These results suggest that this combination induced HA22T/VGH cell death by interfering with redox state regulation by a reduction in pentose phosphate pathway activity and increasing oxidative and nitrosative stress.


Ultrasound in Medicine and Biology | 2008

EFFECTS OF PULSED LOW-INTENSITY ULTRASOUND ON HUMAN CHILD CHONDROCYTES

Yin-Chun Tien; Sin-Daw Lin; Chung-Hwan Chen; Cheng-Chang Lu; Shu-Jem Su; Tsai-Tung Chih

The effect of pulsed low-intensity ultrasound (PLIUS) on human articular chondrocytes was evaluated in an in vitro 3-D agarose gel culture model. Chondrocytes isolated from young childrens articular cartilage of ablated polydactylia were embedded in gel after expansion and exposed to PLIUS on the third day after embedding. Another group of cells was exposed to sham PLIUS as a control. Different intensities of PLIUS treatment-18 mW/cm(2), 48 mW/cm(2), 72 mW/cm(2) and 98 mW/cm(2) (1.0 MHz, with burst duration of 200 micros repeated at 1.0 kHz)-were administered for 20 min/d, and the medium was replaced twice a week. The cultures were evaluated for aggrecan synthesis by enzyme-linked immunosorbent assay (ELISA), type II collagen production by Western blotting or ELISA and cell proliferation by total DNA measurement. The PLIUS was found to increase aggrecan synthesis in a time-dependent manner. The maximal response was observed at an intensity of 48 mW/cm(2). After 14 d of exposure at this intensity, the aggrecan synthesis was 214 +/- 26% of control, and type II collagen synthesis was 148.5 +/- 8.0% of control. However, PLIUS treatment revealed no significant influence on cell proliferation, confirming that the stimulation of aggrecan and type II collagen synthesis by PLIUS was not the result of an increase in chondrocyte cell proliferation. In addition, it was found that human chondrocytes harvested from older donors become less responsive to PLIUS. From this in vitro 3-D study of cultured human chondrocytes, our findings suggest that PLIUS may be applied to the tissue engineering of cartilage constructs.


Toxicology in Vitro | 2013

Caffeine inhibits adipogenic differentiation of primary adipose-derived stem cells and bone marrow stromal cells.

Shu-Hui Su; Huey-Wen Shyu; Yao-Tsung Yeh; Kuan-Ming Chen; Hua Yeh; Shu-Jem Su

Caffeine consumption has been related to loss of body weight and modulates lipid metabolism. However, impacts of caffeine on adipogenic differentiation have not been well determined yet. The present study evaluated the effects of caffeine on adipogenesis using primary rat adipose-derived stem cells (ADSCs) and a mouse bone marrow stromal cell line (M2-10B4) in vitro. ADSCs and M2-10B4 were continuously exposed to caffeine (0.1-1mM) during adipogenic differentiation for 7 and 12 days, respectively. Oil red O and Nile red staining showed that caffeine reduced lipid droplet and adipocyte levels in both cell types. In addition, Nile red staining and FACScan flow cytometry showed that caffeine dose-dependently decreased adipocyte differentiation from 20% to 50% of the control ADSCs and M2-10B4 cells. Caffeine decreased the expression of adipogenesis-related genes including peroxisome proliferator-activated receptor-γ, CCAAT/enhancer-binding protein-α, adipocyte lipid binding protein, lipoprotein lipase, leptin, and TNFα in a dose-dependent manner. Rather, low concentration of caffeine (0.1mM) significantly increased IL-6 expression, but unexpectedly inhibited that at a concentration more than 0.3mM. Taken together, caffeine was able to effectively inhibit adipogenic differentiation of ADSCs and M2-10B4 cells partly through its inhibition of adipogenesis-related factors.


International Journal of Food Sciences and Nutrition | 2013

Caffeine regulates osteogenic differentiation and mineralization of primary adipose-derived stem cells and a bone marrow stromal cell line

Shu-Jem Su; Kee-Lung Chang; Shu-Hui Su; Yao-Tsung Yeh; Huey-Wen Shyu; Kuan-Ming Chen

Caffeine consumption reportedly influences bone mineral density and body weight. However, the effects of caffeine on bone metabolism are still controversial, and whether the dosage of caffeine influences osteogenic differentiation is yet to be clarified. In the present study, we cultured primary adipose-derived stem cells (ADSCs) and a bone marrow stromal cell line (M2-10B4) in osteogenic differentiation media containing varying concentrations of caffeine. Caffeine had biphasic effects: 0.1 mM caffeine significantly enhanced mineralization and alkaline phosphatase (ALP) activity. Consistent with these observations, a caffeine concentration of 0.1 mM upregulated the osteogenic differentiation marker genes ALP and osteocalcin (OCN), and elevated osteoprotegerin (OPG), Runt-related transcription factor 2 (RUNX2) and Sirtuin 1 (SIRT1) levels. However, a concentration of caffeine greater than 0.3 mM suppressed the differentiation of both the cell types. These findings indicate that caffeine has a beneficial effect on ADSCs and bone marrow stromal cells, enhancing differentiation to osteoblasts; this effect, which is mediated via RUNX2 activation at low doses is significantly suppressed at high doses.

Collaboration


Dive into the Shu-Jem Su's collaboration.

Top Co-Authors

Avatar

Kee-Lung Chang

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li-Wen Huang

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bau-Shan Hsieh

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Hsiao-Ling Cheng

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Yu-Chen Hu

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kuan-Ming Chen

Kaohsiung Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge