Network


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

Hotspot


Dive into the research topics where Shi-Jye Chu is active.

Publication


Featured researches published by Shi-Jye Chu.


Free Radical Biology and Medicine | 2014

Stanniocalcin-1 ameliorates lipopolysaccharide-induced pulmonary oxidative stress, inflammation, and apoptosis in mice.

Shih-En Tang; Chin-Pyng Wu; Shu-Yu Wu; Chung-Kan Peng; Wann-Cherng Perng; Bor-Hwang Kang; Shi-Jye Chu; Kun-Lun Huang

Stanniocalcin-1 (STC1) is an endogenous glycoprotein whose anti-inflammatory effects occur through induction of uncoupling proteins to reduce oxidative stress. In this study, we tested the hypothesis that exogenous recombinant human STC1 (rhSTC1) protects against lipopolysaccharide (LPS)-induced acute lung injury in mice. Anesthetized C57BL/6 mice underwent intratracheal spraying of LPS (20 µg/10 g body wt), and lung injury was assessed 24h later by analyzing pulmonary edema, bronchoalveolar lavage fluid, and lung histopathology. Lung inflammation, oxidative stress, and expression of STC1 and its downstream uncoupling protein 2 (UCP2) were analyzed at specific time points. Expression of UCP2 was suppressed initially but was subsequently upregulated after STC1 elevation in response to intratracheal administration of LPS. Intratracheal rhSTC1 treatment 1h before or after LPS spraying significantly attenuated pulmonary inflammation, oxidative stress, cell apoptosis, and acute lung injury. Pretreatment with STC1 short interfering RNA 48 h before LPS spraying inhibited the expression of STC1 and UCP2 and significantly increased the extent of lung injury. These findings suggest that STC1 is an endogenous stress protein that may counteract LPS-induced lung injury by inhibiting the inflammatory cascade and inducing antioxidant and antiapoptotic mechanisms. However, the potential clinical application of STC1 and the direct linkage between UCP2 and LPS-induced lung injury remain to be further investigated.


PLOS ONE | 2013

Protective Effect of Hypercapnic Acidosis in Ischemia-Reperfusion Lung Injury Is Attributable to Upregulation of Heme Oxygenase-1

Shu-Yu Wu; Min-Hui Li; Fu-Chang Ko; Geng-Chin Wu; Kun-Lun Huang; Shi-Jye Chu

Hypercapnic acidosis (HCA) has protective effects in animal models of acute lung injury, but the mechanism underlying the effect of HCA is unclear. Heme oxygenase-1 (HO-1) is an antioxidant enzyme that protects tissue from inflammation injury. We investigated whether HO-1 contributes to the protective effects of HCA in ischemia-reperfusion (IR)-induced lung injury. Typical acute lung injury in rats was successfully induced by 40 min of ischemia and 90 min of reperfusion in an isolated perfused lung model. The rat lungs were randomly assigned to the control group, IR group or IR + HCA group with or without zinc protoporphyrin IX (ZnPP), an HO-1 activity inhibitor. At the end of the experiment, bronchoalveolar lavage fluid (BALF) and lung tissues were collected to evaluate the degree of lung injury. In in vitro experiments, HO-1 siRNA transfected A549 cells were exposed to a normoxic or hypoxia-reoxygenation (H/R) environment in the presence or absence of HCA. IR caused significant increases in the pulmonary arterial pressure, lung weight to body weight and wet/dry ratios, lung weight gain, capillary filtration coefficient, lung injury scores, neutrophil infiltration, and concentrations of protein and TNF-α in the BALF. IR also induced degradation of inhibitor of nuclear factor (NF)-κB-α, increased IκB kinase (IKK)-β phosphorylation and nuclear translocation of NF-κB, and up-regulated HO-1 expression and activity. Furthermore, IR decreased Bcl-2 protein expression and increased the number of active caspase-3 stained cells. HCA treatment enhanced HO-1 expression and activity, and accordingly reduced IKK-NF-κB signaling, inhibited apoptosis, and significantly attenuated IR-induced changes. Treatment with ZnPP partially blocked the protective effect of HCA. In addition, HO-1 siRNA significantly reversed HCA-mediated inhibition of NF-κB signaling in A549 cells subjected to H/R. In conclusion, the protective effect of HCA in IR lung injury in rats was mediated in part by the anti-inflammatory and anti-apoptotic action of HO-1.


Anesthesiology | 2015

Valproic Acid Attenuates Acute Lung Injury Induced by Ischemia–reperfusion in Rats

Shu-Yu Wu; Shih-En Tang; Fu-Chang Ko; Geng-Chin Wu; Kun-Lun Huang; Shi-Jye Chu

Background:Evidence reveals that histone deacetylase (HDAC) inhibition has potential for the treatment of inflammatory diseases. The protective effect of HDAC inhibition involves multiple mechanisms. Heme oxygenase-1 (HO-1) is protective in lung injury as a key regulator of antioxidant response. The authors examined whether HDAC inhibition provided protection against ischemia–reperfusion (I/R) lung injury in rats by up-regulating HO-1 activity. Methods:Acute lung injury was induced by producing 40 min of ischemia followed by 60 min of reperfusion in isolated perfused rat lungs. The rats were randomly allotted to control group, I/R group, or I/R + valproic acid (VPA) group with or without an HO-1 activity inhibitor (zinc protoporphyrin IX) (n = 6 per group). Results:I/R caused significant increases in the lung edema, pulmonary arterial pressure, lung injury scores, tumor necrosis factor-&agr;, and cytokine-induced neutrophil chemoattractant-1 concentrations in bronchoalveolar lavage fluid. Malondialdehyde levels, carbonyl contents, and myeloperoxidase-positive cells in lung tissue were also significantly increased. I/R stimulated the degradation of inhibitor of nuclear factor-&kgr;B-&agr;, nuclear translocation of nuclear factor-&kgr;B, and up-regulation of HO-1 activity. Furthermore, I/R decreased B-cell lymphoma-2, heat shock protein 70, acetylated histone H3 protein expression, and increased the caspase-3 activity in the rat lungs. In contrast, VPA treatment significantly attenuated all the parameters of lung injury, oxidative stress, apoptosis, and inflammation. In addition, VPA treatment also enhanced HO-1 activity. Treatment with zinc protoporphyrin IX blocked the protective effect of VPA. Conclusions:VPA protected against I/R-induced lung injury. The protective mechanism may be partly due to enhanced HO-1 activity following HDAC inhibition.


Shock | 2004

Effect of hyperbaric oxygen on endotoxin-induced lung injury in rats

Wann-Cherng Perng; Chin-Pyng Wu; Shi-Jye Chu; Bor-Hwang Kang; Kun-Lun Huang

Oxygen therapy remains the main component of the ventilation strategy for treatment of patients with acute lung injury. Hyperbaric oxygen therapy (HBO2) is the intermittent administration of 100% oxygen at pressure greater than sea level and has been applied widely to alleviate a variety of hypoxia-related tissue injuries. The purpose of this study was to evaluate the effect of hyperbaric oxygen on acute lung injury induced by intratracheal spraying of lipopolysaccharide (LPS) in rats. Male Sprague-Dawley rats underwent implantation of a carotid artery catheter under general anesthesia. Aerosolized LPS was delivered twice into the lungs via intratracheal puncture. Animals were either breathing room air (n = 27) or subjected to hyperbaric oxygen (HBO2) exposure (n = 27) 1 h after LPS spraying. Acute lung injury was evaluated 5 h and 24 h later. Compared with the control group, intratracheal spraying of LPS caused profound hypoxemia, greater wet/dry weight ratio (W/D) of the lung (5.67 ± 0.22 vs. 4.98 ± 0.19), and higher protein concentration (1706 ± 168 vs. 200 ± 90 mg/L) and LDH activity (129 ± 30 vs. 46 ± 15, mAbs/min) in bronchoalveolar lavage (BAL) fluid. Intratracheal spraying of LPS also caused significant WBC sequestration in the lung tissue. HBO2 treatment significantly reverted hypoxemia, reduced lung injury measures evaluated at 5 and 24 h, and enhanced 24-h animal survival rate (χ2 = 5.08, P = 0.024). The malondialdehyde (MDA) concentrations in lung tissue and serum were both increased after LPS spraying. Neither single HBO2 therapy nor five sequential daily treatments enhanced MDA production in lung tissue or serum. Our results suggested that hyperbaric oxygen might reduce acute lung injury caused by intratracheal spraying of LPS in rats. This treatment modality is not associated with enhancement of oxidative stress to the lung.


Wound Repair and Regeneration | 2008

Hyperbaric oxygen attenuates cell growth in skin fibroblasts cultured in a high-glucose medium

Hen-I Lin; Shi-Jye Chu; Wann-Cherng Perng; Chin-Pyng Wu; Zuei-Yin Lin; Kun-Lun Huang

Hyperglycemia and hypoxia synergistically retard diabetic wound healing. We investigated the direct effect of hyperbaric and normobaric hyperoxia on skin fibroblasts cultured in a high‐glucose medium. Detroit 551 human dermal fibroblasts cultured in Dulbeccos modified Eagles medium containing d‐glucose had reduced cell survival compared with cells grown in normal glucose medium; survival was 27.5±3.8% lower in 25 mM glucose and 30.6±3.7% lower in 50 mM glucose. Cell survival decreased because of inhibition of cell proliferation and enhanced cell death. Daily hyperbaric oxygen therapy at 2.5 atmosphere absolute for 90 minutes on 3 consecutive days reduced cell proliferation and increased cell death in normal cultured fibroblasts. Hyperbaric oxygen therapy and high‐glucose medium had a synergistic effect and reduced survival by 37.6±4.4% (25 mM glucose) and 39.6±5.1% (50 mM glucose). The effects of hyperbaric oxygen and high‐glucose medium were associated with overproduction of reactive oxygen species. Our results suggest that direct exposure of skin fibroblasts to hyperbaric oxygen affects cell growth and superimposes the toxic effect of high glucose. This cytotoxicity may be related to the production of reactive oxygen species in the fibroblasts.


International Journal of Molecular Sciences | 2017

Ac2-26, an Annexin A1 Peptide, Attenuates Ischemia-Reperfusion-Induced Acute Lung Injury

Wen-I Liao; Shu-Yu Wu; Geng-Chin Wu; Hsin-Ping Pao; Shih-En Tang; Kun-Lun Huang; Shi-Jye Chu

Annexin A1 (AnxA1) is an endogenous protein that modulates anti-inflammatory processes, and its therapeutic potential has been reported in a range of inflammatory diseases. The effect of AnxA1 on ischemia-reperfusion (IR)-induced lung injury has not been examined. In this study, isolated, perfused rat lungs were subjected to IR lung injury induced by ischemia for 40 min, followed by reperfusion for 60 min. The rat lungs were randomly treated with vehicle (phosphate-buffered saline), and Ac2-26 (an active N-terminal peptide of AnxA1) with or without an N-formyl peptide receptor (FPR) antagonist N-Boc-Phe-Leu-Phe-Leu-Phe (Boc2). An in vitro study of the effects of Ac2-26 on human alveolar epithelial cells subjected to hypoxia-reoxygenation was also investigated. Administration of Ac2-26 in IR lung injury produced a significant attenuation of lung edema, pro-inflammatory cytokine production recovered in bronchoalveolar lavage fluid, oxidative stress, apoptosis, neutrophil infiltration, and lung tissue injury. Ac2-26 also decreased AnxA1 protein expression, inhibited the activation of nuclear factor-κB and mitogen-activated protein kinase pathways in the injured lung tissue. Finally, treatment with Boc2 abolished the protective action of Ac2-26. The results indicated that Ac2-26 had a protective effect against acute lung injury induced by IR, which may be via the activation of the FPR.


International Immunopharmacology | 2015

Protection against reperfusion lung injury via aborgating multiple signaling cascades by trichostatin A

Hsian-He Hsu; Shu-Yu Wu; Shih-En Tang; Geng-Chin Wu; Min-Hui Li; Kun-Lun Huang; Shi-Jye Chu

Trichostatin A (TSA) is a histone deacetylase inhibitor with anti-inflammatory effects. Nonetheless, little information is available about the effect of TSA in ischemia-reperfusion (IR)-induced lung injury. In a perfused rat lung model, IR was induced by 40min of ischemia followed by 60min of reperfusion. The rat lungs were randomly divided into several groups including control, control+TSA (0.1mg/kg), IR, and IR+various dosages of TSA (0.05, 0.075, 0.1mg/kg). Bronchoalveolar lavage fluids and lung tissues were obtained and examined at the end of the experiment. TSA dose-dependently diminished IR-induced increased vascular permeability and edema, pulmonary artery pressure, and histological changes in the lungs. Additionally, TSA suppressed lavage tumor necrosis factor-α and cytokine-induced neutrophil chemoattractant concentrations, cell infiltration, and myeloperoxidase-positive cells in the lung tissue. Furthermore, TSA attenuated the phosphorylation of extracellular signal-regulated kinase, p38, and c-Jun N-terminal kinase, degradation of the inhibitor of nuclear factor (NF)-κB, and nuclear NF-κB levels. TSA also decreased poly (ADP-ribose) polymerase but enhanced acetylated histone H3 acetylation, Bcl-2, and mitogen-activated protein kinase phosphatase-1 (MKP-1) expression in IR lung tissue. Therefore, TSA exerted a protective effect on IR-induced lung injury via increasing histone acetylation and MKP-1 protein expression, repressing NF-κB, mitogen-activated protein kinase, and apoptosis signaling pathways.


Inflammation | 2017

Protective Effects of Neural Crest-Derived Stem Cell-Conditioned Media against Ischemia-Reperfusion-Induced Lung Injury in Rats

Chung-Kan Peng; Shu-Yu Wu; Shih-En Tang; Min-Hui Li; Shih-Shiuan Lin; Shi-Jye Chu; Kun-Lun Huang

Current treatments for ischemia-reperfusion (IR)-induced acute lung injury are limited. Mesenchymal stem cell-conditioned medium (CM) has been reported to attenuate lung injury. Neural crest stem cells (NCSCs), a type of multipotent stem cells, are more easily obtained than mesenchymal stem cells. We hypothesize that NCSC-CM has anti-inflammatory properties that could protect against IR-induced lung injury in rats. In this study, NCSC-CM was derived from rat NCSCs. Typical acute lung injury was induced by 30-min ischemia followed by 90-min reperfusion in adult male Sprague–Dawley rats. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected to analyze the degree of lung injury after the experiment. NCSC-CM was administered before ischemia and after reperfusion. NCSC-CM treatment significantly attenuated IR-induced lung edema, as indicated by decreases in pulmonary vascular permeability, lung weight gain, wet to dry weight ratio, lung weight to body weight ratio, pulmonary arterial pressure, and protein level in BALF. The levels of tumor necrosis factor-α and interleukin-6 in the BALF were also significantly decreased. Additionally, NCSC-CM improved lung pathology and neutrophil infiltration in the lung tissue, and significantly suppressed nuclear factor (NF)-κB activity and IκB-α degradation in the lung. However, heating NCSC-CM eliminated these protective effects. Our experiment demonstrates that NCSC-CM treatment decreases IR-induced acute lung injury and that the protective mechanism may be attributable to the inhibition of NF-κB activation and the inflammatory response. Therefore, NCSC-CM may be a novel approach for treating IR-induced lung injury.


International Journal of Molecular Sciences | 2018

Correction: Wen-I Liao, et al. Ac2-26, an Annexin A1 Peptide, Attenuates Ischemia-Reperfusion-Induced Acute Lung Injury. Int. J. Mol. Sci. 2017, 18, 1771

Wen-I Liao; Shu-Yu Wu; Geng-Chin Wu; Hsin-Ping Pao; Shih-En Tang; Kun-Lun Huang; Shi-Jye Chu

The authors would like to make a correction to their published paper [1][...].


重症醫學雜誌 | 2010

Nosocomial Urinary Tract Infection Related Retroperitoneal Abscess with Internal Colonic Fistula

Shih-Hung Tsai; Wei-Chou Chang; Chin-Wang Hsu; Shi-Jye Chu; Wann-Cherng Perng

Retroperitoneal abscess may not be diagnosed timely occasionally. Here we report a patient who had nosocomial urinary tract infection related retroperitoneal abscess and subsequently developed a secondary internal fistula to the adjacent descending colon. Nosocomial UTI related retroperitoneal abscess can be a source of occult infection that responses poorly to antibiotic treatment. Retroperitoneal abscess can be further complicated with internal colonic fistula. Repeated fistulography and culture may be necessary to identify the extensiveness of the abscess cavity and the development of resistant strains. The presence of multiple drug resistance pathogens can perplex the treatment course and repeated drainage will be required.

Collaboration


Dive into the Shi-Jye Chu's collaboration.

Top Co-Authors

Avatar

Kun-Lun Huang

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shu-Yu Wu

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Geng-Chin Wu

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shih-En Tang

Tri-Service General Hospital

View shared research outputs
Top Co-Authors

Avatar

Chin-Pyng Wu

Tri-Service General Hospital

View shared research outputs
Top Co-Authors

Avatar

Min-Hui Li

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chung-Kan Peng

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hsin-Ping Pao

National Defense Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wann-Cherng Perng

Tri-Service General Hospital

View shared research outputs
Top Co-Authors

Avatar

Wen-I Liao

Tri-Service General Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge