Network


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

Hotspot


Dive into the research topics where Gong-Jhe Wu is active.

Publication


Featured researches published by Gong-Jhe Wu.


Toxicology and Applied Pharmacology | 2008

Ketamine inhibits tumor necrosis factor-α and interleukin-6 gene expressions in lipopolysaccharide-stimulated macrophages through suppression of toll-like receptor 4-mediated c-Jun N-terminal kinase phosphorylation and activator protein-1 activation

Gong-Jhe Wu; Ta-Liang Chen; Yune-Fang Ueng; Ruei-Ming Chen

Our previous study showed that ketamine, an intravenous anesthetic agent, has anti-inflammatory effects. In this study, we further evaluated the effects of ketamine on the regulation of tumor necrosis factor-alpha (TNF-alpha) and interlukin-6 (IL-6) gene expressions and its possible signal-transducing mechanisms in lipopolysaccharide (LPS)-activated macrophages. Exposure of macrophages to 1, 10, and 100 microM ketamine, 100 ng/ml LPS, or a combination of ketamine and LPS for 1, 6, and 24 h was not cytotoxic to macrophages. A concentration of 1000 microM of ketamine alone or in combined treatment with LPS caused significant cell death. Administration of LPS increased cellular TNF-alpha and IL-6 protein levels in concentration- and time-dependent manners. Meanwhile, treatment with ketamine concentration- and time-dependently alleviated the enhanced effects. LPS induced TNF-alpha and IL-6 mRNA syntheses. Administration of ketamine at a therapeutic concentration (100 microM) significantly inhibited LPS-induced TNF-alpha and IL-6 mRNA expressions. Application of toll-like receptor 4 (TLR4) small interfering (si)RNA into macrophages decreased cellular TLR4 levels. Co-treatment of macrophages with ketamine and TLR4 siRNA decreased the LPS-induced TNF-alpha and IL-6 productions more than alone administration of TLR4 siRNA. LPS stimulated phosphorylation of c-Jun N-terminal kinase and translocation of c-Jun and c-Fos from the cytoplasm to nuclei. However, administration of ketamine significantly decreased LPS-induced activation of c-Jun N-terminal kinase and translocation of c-Jun and c-Fos. LPS increased the binding of nuclear extracts to activator protein-1 consensus DNA oligonucleotides. Administration of ketamine significantly ameliorated LPS-induced DNA binding activity of activator protein-1. Therefore, a clinically relevant concentration of ketamine can inhibit TNF-alpha and IL-6 gene expressions in LPS-activated macrophages. The suppressive mechanisms occur through suppression of TLR4-mediated sequential activations of c-Jun N-terminal kinase and activator protein-1.


Anesthesiology | 2003

Propofol suppresses macrophage functions and modulates mitochondrial membrane potential and cellular adenosine triphosphate synthesis

Ruei-Ming Chen; Chih-Hsiung Wu; Huai-Chia Chang; Gong-Jhe Wu; Yi Ling Lin; Joen Rong Sheu; Ta-Liang Chen

Background Propofol is an intravenous anesthetic agent that may impair host defense system. The aim of this study was to evaluate the effects of propofol on macrophage functions and its possible mechanism. Methods Mouse macrophage-like Raw 264.7 cells were exposed to propofol, at 3, 30 (a clinically relevant concentration), and 300 &mgr;m. Cell viability, lactate dehydrogenase, and cell cycle were analyzed to determine the cellular toxicity of propofol to macrophages. After administration of propofol, chemotactic, phagocytic, and oxidative ability and interferon-&ggr; mRNA production were carried out to validate the potential effects of propofol on macrophage functions. Mitochondrial membrane potential and cellular adenosine triphosphate levels were also analyzed to evaluate the role of mitochondria in propofol-induced macrophage dysfunction. Results Exposure of macrophages to 3 and 30 &mgr;m propofol did not affect cell viability. When the administered concentration reached 300 &mgr;m, propofol would increase lactate dehydrogenase release, cause arrest of cell cycle in G1/S phase, and lead to cell death. In the 1-h-treated macrophages, propofol significantly reduced macrophage functions of chemotactic and oxidative ability in a concentration-dependent manner. However, the suppressive effects were partially or completely reversed after 6 and 24 h. Propofol could reduce phagocytic activities of macrophages in concentration- and time-dependent manners. Exposure of macrophages to lipopolysaccharide induced the mRNA of interferon-&ggr;, but the induction was significantly blocked by propofol. Propofol concentration-dependently decreased the membrane potential of macrophage mitochondria, but the effects were descended with time. The levels of cellular adenosine triphosphate in macrophages were also reduced by propofol. Conclusions A clinically relevant concentration of propofol can suppress macrophage functions, possibly through inhibiting their mitochondrial membrane potential and adenosine triphosphate synthesis instead of direct cellular toxicity.


Journal of Agricultural and Food Chemistry | 2012

Neuroprotective effects of xanthohumol, a prenylated flavonoid from hops (humulus lupulus), in ischemic stroke of rats

Ting Lin Yen; Chung King Hsu; Wan-Jung Lu; Cheng-ying Hsieh; Duen-Suey Chou; Gong-Jhe Wu; Joen Rong Sheu

Xanthohumol is the principal prenylated flavonoid in hops (Humulus lupulus L.), an ingredient of beer. Xanthohumol was found to be a potent chemopreventive agent; however, no data are available concerning its neuroprotective effects. In the present study, the neuroprotective activity and mechanisms of xanthohumol in rats with middle cerebral artery occlusion (MCAO)-induced cerebral ischemia were examined. Treatment with xanthohumol (0.2 and 0.4 mg/kg; intraperitoneally) 10 min before MCAO dose-dependently attenuated focal cerebral ischemia and improved neurobehavioral deficits in cerebral ischemic rats. Xanthohumol treatment produced a marked reduction in infarct size compared to that in control rats. MCAO-induced focal cerebral ischemia was associated with increases in hypoxia-inducible factor (HIF)-1α, tumor necrosis factor (TNF)-α, inducible nitric oxide synthase (iNOS), and active caspase-3 protein expressions in ischemic regions. These expressions were obviously inhibited by treatment with xanthohumol. In addition, xanthohumol (3-70 μM) concentration-dependently inhibited platelet aggregation stimulated by collagen (1 μg/mL) in human platelet-rich plasma. An electron spin resonance (ESR) method was used to examine the scavenging activity of xanthohumol on free radicals which had formed. Xanthohumol (1.5 and 3 μM) markedly reduced the ESR signal intensity of hydroxyl radical (OH•) formation in the H₂O₂/NaOH/DMSO system. In conclusion, this study demonstrates for the first time that in addition to its originally being considered an agent preventing tumor growth, xanthohumol possesses potent neuroprotective activity. This activity is mediated, at least in part, by inhibition of inflammatory responses (i.e., HIF-1α, iNOS expression, and free radical formation), apoptosis (i.e., TNF-α, active caspase-3), and platelet activation, resulting in a reduction of infarct volume and improvement in neurobehavior in rats with cerebral ischemia. Therefore, this novel role of xanthohumol may represent high therapeutic potential for treatment or prevention of ischemia-reperfusion injury-related disorders.


Annals of the New York Academy of Sciences | 2005

Propofol specifically inhibits mitochondrial membrane potential but not complex I NADH dehydrogenase activity, thus reducing cellular ATP biosynthesis and migration of macrophages

Gong-Jhe Wu; Yu-Ting Tai; Ta-Liang Chen; Li Ling Lin; Yune-Fang Ueng; Ruei-Ming Chen

Abstract: Propofol is a widely used intravenous anesthetic agent. Our previous study showed that a therapeutic concentration of propofol can modulate macrophage functions. Mitochondria play critical roles in the maintenance of macrophage activities. This study attempted to evaluate further the effects of mitochondria on the propofol‐induced suppression of macrophage functions using mouse macrophage‐like Raw 264.7 cells as the experimental model. Macrophages were exposed to a clinically relevant concentration of propofol for 1, 6, and 24 h. Analysis by the Trypan blue exclusion method revealed that propofol was not cytotoxic to macrophages. Exposure of macrophages to propofol did not affect mitochondrial NADH dehydrogenase activity of complex I. However, analysis of flow cytometry showed that propofol significantly decreased the mitochondrial membrane potential of macrophages. Cellular levels of ATP in macrophages were significantly reduced after propofol administration. In parallel with the dysfunction of mitochondria, the chemotactic analysis showed that exposure to propofol significantly inhibited the migration of macrophages. This study shows that a therapeutic concentration of propofol can specifically reduce the mitochondrial membrane potential, but there is no such effect on complex I NADH dehydrogenase activity. Modulation of the mitochondrial membrane potential may decrease the biosynthesis of cellular ATP and thus reduce the chemotactic activity of macrophages. This study provides in vitro data to validate mitochondrial dysfunction as a possible critical cause for propofol‐induced immunosuppression of macrophage functions.


Chemico-Biological Interactions | 2009

Propofol suppresses tumor necrosis factor-α biosynthesis in lipopolysaccharide-stimulated macrophages possibly through downregulation of nuclear factor-kappa B-mediated toll-like receptor 4 gene expression

Gong-Jhe Wu; Ta-Liang Chen; Chia Chen Chang; Ruei-Ming Chen

Lipopolysaccharide (LPS), a gram-negative bacterial outer membrane component, can activate macrophages via a toll-like receptor 4-dependent pathway. Our previous study has shown that propofol, an intravenous anesthetic reagent, has anti-inflammatory effects. This study was further aimed to evaluate the roles of toll-like receptor 4 in propofol-caused suppression of tumor necrosis factor-alpha (TNF-alpha) biosynthesis in LPS-stimulated macrophages and its possible molecular mechanisms. Exposure of macrophages to propofol and LPS did not affect cell viability. Meanwhile, the LPS-caused augmentations in the productions of TNF-alpha protein and mRNA were significantly decreased following incubation with a therapeutic concentration of propofol (50 microM). Analysis of toll-like receptor 4 small interference (si)RNA revealed that this membrane receptor might participate in the propofol-caused suppression of TNF-alpha biosynthesis. Treatment of macrophages with LPS-induced toll-like receptor 4 protein and mRNA productions. Propofol at a clinically relevant concentration could inhibit such induction. In parallel, the LPS-induced translocation and transactivation of transcription factor nuclear factor-kappa B (NFkappaB) were significantly alleviated following propofol incubation. There are several NFkappaB DNA-binding motifs found in the promoter region of toll-like receptor 4. Therefore, this study shows that propofol at a therapeutic concentration can downregulate TNF-alpha biosynthesis possibly via inhibition of NFkappaB-mediated toll-like receptor 4 gene expression.


Annals of the New York Academy of Sciences | 2005

2,6-Diisopropylphenol protects osteoblasts from oxidative stress-induced apoptosis through suppression of caspase-3 activation.

Ruei-Ming Chen; Gong-Jhe Wu; Huai-Chia Chang; Jue Tai Chen; Tzeng-Fu Chen; Yi Ling Lin; Ta-Liang Chen

Abstract: 2,6‐Diisopropylphenol is an intravenous anesthetic agent used for induction and maintenance of anesthesia. Since it is similar to α‐tocopherol, 2,6‐diisopropylphenol may have antioxidant effects. Osteoblasts play important roles in bone remodeling. In this study, we attempted to evaluate the protective effects of 2,6‐diisopropylphenol on oxidative stress‐induced osteoblast insults and their possible mechanisms, using neonatal rat calvarial osteoblasts as the experimental model. Clinically relevant concentrations of 2,6‐diisopropylphenol (3 and 30 μM) had no effect on osteoblast viability. However, 2,6‐diisopropylphenol at 300 μM time‐dependently caused osteoblast death. Exposure to sodium nitroprusside (SNP), a nitric oxide donor, increased amounts of nitrite in osteoblasts. 2,6‐Diisopropylphenol did not scavenge basal or SNP‐releasing nitric oxide. Hydrogen peroxide (HP) enhanced levels of intracellular reactive oxygen species in osteoblasts. 2,6‐Diisopropylphenol significantly reduced HP‐induced oxidative stress. Exposure of osteoblasts to SNP and HP decreased cell viability time‐dependently. 2,6‐Diisopropylphenol protected osteoblasts from SNP‐ and HP‐induced cell damage. Analysis by a flow cytometric method revealed that SNP and HP induced osteoblast apoptosis. 2,6‐Diisopropylphenol significantly blocked SNP‐ and HP‐induced osteoblast apoptosis. Administration of SNP and HP increased caspase‐3 activities. However, 2,6‐diisopropylphenol significantly decreased SNP‐ and HP‐enhanced caspase‐3 activities. This study shows that a therapeutic concentration of 2,6‐diisopropylphenol can protect osteoblasts from SNP‐ and HP‐induced cell insults, possibly via suppression of caspase‐3 activities.


Toxicology and Applied Pharmacology | 2016

Honokiol induces autophagic cell death in malignant glioma through reactive oxygen species-mediated regulation of the p53/PI3K/Akt/mTOR signaling pathway

Chien-Ju Lin; Ta-Liang Chen; Yuan-Yun Tseng; Gong-Jhe Wu; Ming-Hui Hsieh; Yung-Wei Lin; Ruei-Ming Chen

Honokiol, an active constituent extracted from the bark of Magnolia officinalis, possesses anticancer effects. Apoptosis is classified as type I programmed cell death, while autophagy is type II programmed cell death. We previously proved that honokiol induces cell cycle arrest and apoptosis of U87 MG glioma cells. Subsequently in this study, we evaluated the effect of honokiol on autophagy of glioma cells and examined the molecular mechanisms. Administration of honokiol to mice with an intracranial glioma increased expressions of cleaved caspase 3 and light chain 3 (LC3)-II. Exposure of U87 MG cells to honokiol also induced autophagy in concentration- and time-dependent manners. Results from the addition of 3-methyladenine, an autophagy inhibitor, and rapamycin, an autophagy inducer confirmed that honokiol-induced autophagy contributed to cell death. Honokiol decreased protein levels of PI3K, phosphorylated (p)-Akt, and p-mammalian target of rapamycin (mTOR) in vitro and in vivo. Pretreatment with a p53 inhibitor or transfection with p53 small interfering (si)RNA suppressed honokiol-induced autophagy by reversing downregulation of p-Akt and p-mTOR expressions. In addition, honokiol caused generation of reactive oxygen species (ROS), which was suppressed by the antioxidant, vitamin C. Vitamin C also inhibited honokiol-induced autophagic and apoptotic cell death. Concurrently, honokiol-induced alterations in levels of p-p53, p53, p-Akt, and p-mTOR were attenuated following vitamin C administration. Taken together, our data indicated that honokiol induced ROS-mediated autophagic cell death through regulating the p53/PI3K/Akt/mTOR signaling pathway.


Surgery | 2013

Propofol protects against nitrosative stress-induced apoptotic insults to cerebrovascular endothelial cells via an intrinsic mitochondrial mechanism

Ruei-Ming Chen; Yu-Tyng Tai; Tyng-Guey Chen; The-Hin Lin; Huai-Chia Chang; Ta-Liang Chen; Gong-Jhe Wu

BACKGROUND Cerebrovascular endothelial cells (CECs), major component cells of the blood-brain barrier, can be injured by oxidative stress. Propofol can protect cells from oxidative injury. The aim of this study was to evaluate the effects of propofol on nitrosative stress-induced insults to CECs and its possible mechanisms. METHODS Primary CECs isolated from mouse cerebral capillaries were exposed to2 nitric oxide (NO) donors: sodium nitroprusside (SNP) or S-nitrosoglutathione (GSNO). Cellular NO levels, cell morphologies, and cell viabilities were analyzed. DNA fragmentation and apoptotic cells were quantified using flow cytometry. Proapoptotic Bcl2-antagonist-killer (Bak) and cytochrome c were immunodetected. Bak translocation was analyzed using confocal microscopy. Caspases-9 and -3 activities were measured fluorometrically. Permeability of the CEC monolayer was assayed by measuring the transendothelial electrical resistance. RESULTS Exposure of CECs to SNP increased cellular NO levels and simultaneously decreased cell viability (P < .01). Meanwhile, treatment of CECs with propofol at a therapeutic concentration (50 μM) decreased SNP-induced cell death (P < .01). SNP induced DNA fragmentation and cell apoptosis, but propofol decreased the cell injury (P < .01). Sequentially, propofol decreased SNP-enhanced Bak levels and translocation from the cytoplasm to mitochondria (P < .05). Exposure of CECs to propofol attenuated GSNO-induced cell death, apoptosis, and caspase-3 activation (P < .01). Additionally, propofol protected CECs against SNP-induced disruption of the CEC monolayer (P < .05). Consequently, SNP-enhanced cascade activation of caspases-9 and -3 was decreased by propofol (P < .01). CONCLUSION This study suggested that propofol at a therapeutic concentration can protect against nitrosative stress-induced apoptosis of CECs due to downregulation of the intrinsic Bak-mitochondrion-cytochrome c-caspase protease pathway.


Chemico-Biological Interactions | 2010

Nitrosative stress induces osteoblast apoptosis through downregulating MAPK-mediated NFκB/AP-1 activation and subsequent Bcl-XL expression

Ta-Liang Chen; Gong-Jhe Wu; Chun-Sen Hsu; Tsorng-Harn Fong; Ruei-Ming Chen

During inflammation, a large amount of reactive oxygen species is produced and causes insults to osteoblasts. This study was aimed to evaluate the molecular mechanisms of sodium nitroprusside (SNP)-induced insults to rat osteoblasts. Exposure of osteoblasts, prepared from neonatal rat calvaria to SNP increased the levels of cellular nitric oxide and intracellular reactive oxygen species, and simultaneously induced apoptotic insults in concentration- or time-dependent manners. Exposure of rat osteoblasts to SNP time-dependently decreased antiapoptotic Bcl-X(L) messenger RNA and protein syntheses. Treatment of rat osteoblasts with SNP decreased the translocation of transcription factors nuclear factor-kappaB (NFkappaB) and activator protein (AP)-1 from the cytoplasm to nuclei. Sequentially, phosphorylations of the mitogen-activated protein kinases (MAPKs) of ERK1/2, JNK1/2, and p38 MAPK decreased following SNP administration. Application of ERK1 and JNK1 small interference (si)RNAs into rat osteoblasts decreased the translation of these MAPKs and synergistically enlarged SNP-caused alterations in Bcl-X(L) mRNA expression and cell apoptosis. Therefore, this study shows that the SNP-induced nitrosative stress decreased Bcl-X(L) expression, and then induced apoptotic insults to rat osteoblasts through downregulating phosphorylation of MAPKs and subsequent activation of NFkappaB and AP-1.


European Journal of Pharmacology | 2010

Inhibitory signaling of 17β-estradiol in platelet activation: The pivotal role of cyclic AMP-mediated nitric oxide synthase activation

Gong-Jhe Wu; Jie Jen Lee; Duen-Suey Chou; Thanasekaran Jayakumar; Wei Fan Chen; Joen Rong Sheu

Arterial thromboses are mostly composed of platelets adherent to ruptured endothelial surfaces. Platelets are anucleated cells; therefore, they represent an excellent and unique model to selectively investigate the signaling pathways mediating the nongenomic effects of estrogen. The aim of this study was to examine the signal transduction pathways of 17β-estradiol in preventing platelet activation. In this study, 17β-estradiol (5~10 μM) exhibited more-potent activity of inhibiting platelet aggregation stimulated by collagen than other agonists (i.e., thrombin). 17β-Estradiol-inhibited collagen-stimulated platelet activation accompanied by [Ca(2+)]i mobilization, thromboxane A₂ (TxA₂) formation, and phospholipase C (PLC)γ2, protein kinase C (PKC), and p38 mitogen-activated protein kinase (MAPK) phosphorylation. 17β-Estradiol markedly increased cyclic AMP and cyclic GMP levels, nitric oxide (NO) release, vasodilator-stimulated phosphoprotein (VASP) phosphorylation, and endothelial nitric oxide synthase (eNOS) expression. SQ 22536, an inhibitor of adenylate cyclase, markedly reversed the 17β-estradiol-mediated effects (i.e., platelet aggregation, and PLCγ2, VASP, and eNOS phosphorylation). Furthermore, ICI 182,780, a pure estrogen receptor antagonist, also reversed the 17β-estradiol-mediated effects on platelet aggregation and eNOS activation. In conclusion, the most important findings of this study demonstrate for the first time that the inhibitory effect of 17β-estradiol in platelet activation involves activation of the cyclic AMP-eNOS/NO-cyclic GMP pathway, resulting in inhibition of PLCγ2 and p38 MAPK activation, which may lower the incidence of cardiovascular events in postmenopausal women.

Collaboration


Dive into the Gong-Jhe Wu's collaboration.

Top Co-Authors

Avatar

Ruei-Ming Chen

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

Ta-Liang Chen

Taipei Medical University Hospital

View shared research outputs
Top Co-Authors

Avatar

Chi-Sheng Hung

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Hung-Wei Chang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Huai-Chia Chang

Taipei Medical University Hospital

View shared research outputs
Top Co-Authors

Avatar

Yi Ling Lin

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

F. Y. Huang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Joen Rong Sheu

Taipei Medical University

View shared research outputs
Top Co-Authors

Avatar

W. L. Peng

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Yu-Ting Tai

Taipei Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge