Network


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

Hotspot


Dive into the research topics where Xiance Sun is active.

Publication


Featured researches published by Xiance Sun.


Chemico-Biological Interactions | 2010

Genotoxic effect of 6-gingerol on human hepatoma G2 cells

Guang Yang; Laifu Zhong; Liping Jiang; Chengyan Geng; Jun Cao; Xiance Sun; Yufang Ma

6-gingerol, a major component of ginger, has antioxidant, anti-apoptotic, and anti-inflammatory activities. However, some dietary phytochemicals possess pro-oxidant effects as well, and the risk of adverse effects is increased by raising the use of doses. The aim of this study was to assess the genotoxic effects of 6-gingerol and to clarify the mechanisms, using human hepatoma G2 (HepG2) cells. Exposure of the cells to 6-gingerol caused significant increase of DNA migration in comet assay, increase of micronuclei frequencies at high concentrations at 20-80 and 20-40 microM, respectively. These results indicate that 6-gingerol caused DNA strand breaks and chromosome damage. To further elucidate the underlying mechanisms, we tested lysosomal membrane stability, mitochondrial membrane potential, the intracellular generation of reactive oxygen species (ROS) and reduced glutathione (GSH). In addition, the level of oxidative DNA damage was evaluated by immunocytochemical analysis on 8-hydroxydeoxyguanosine (8-OHdG). Results showed that lysosomal membrane stability was reduced after treatment by 6-gingerol (20-80 microM) for 40 min, mitochondrial membrane potential decreased after treatment for 50 min, GSH and ROS levels were significantly increased after treatment for 60 min. These suggest 6-gingerol induces genotoxicity probably by oxidative stress; lysosomal and mitochondrial damage were observed in 6-gingerol-induced toxicity.


Phytotherapy Research | 2011

6-Gingerol Prevents Patulin-induced Genotoxicity in HepG2 Cells

Guang Yang; Laifu Zhong; Liping Jiang; Chengyan Geng; Jun Cao; Xiance Sun; Xiaofang Liu; Min Chen; Yufang Ma

Patulin (PAT) is a mycotoxin produced by several Penicillium, Aspergillus and Byssochlamys species. Since PAT is a potent genotoxic compound, and PAT contamination is common in fruits and fruit products, the search for newer, better agents for protection against genotoxicity of PAT is required. In this study, the chemoprotective effect of 6‐gingerol against PAT‐induced genotoxicity in HepG2 cells was investigated. The comet assay and micronucleus test (MNT) were used to monitor genotoxic effects. To further elucidate the underlying mechanisms, the intracellular generation of reactive oxygen species (ROS) and level of reduced glutathione (GSH) were tested. In addition, the level of oxidative DNA damage was evaluated by immunocytochemical analysis of 8‐hydroxydeoxyguanosine (8‐OHdG). The results showed that 6‐gingerol significantly reduced the DNA strand breaks and micronuclei formation caused by PAT. Moreover, 6‐gingerol effectively suppressed PAT‐induced intracellular ROS formation and 8‐OHdG level. The GSH depletion induced by PAT in HepG2 cells was also attenuated by 6‐gingerol pretreatment. These findings suggest that 6‐gingerol has a strong protective ability against the genotoxicity caused by PAT, and the antioxidant activity of 6‐gingerol may play an important part in attenuating the genotoxicity of PAT. Copyright


Phytotherapy Research | 2012

6-Gingerol induces apoptosis through lysosomal-mitochondrial axis in human hepatoma G2 cells.

Guang Yang; Shaopeng Wang; Laifu Zhong; Xu Dong; Wenli Zhang; Liping Jiang; Chengyan Geng; Xiance Sun; Xiaofang Liu; Min Chen; Yufang Ma

6‐Gingerol, a major phenolic compound derived from ginger, has been known to possess anticarcinogenic activities. However, the mechanisms are not well understood. In our previous study, it was demonstrated that lysosome and mitochondria may be the primary targets for 6‐gingerol in HepG2 cells. Therefore, the aim was to evaluate lysosome‐mitochondria cross‐signaling in 6‐gingerol‐induced apoptosis. Apoptosis was detected by Hoechst 33342 and TUNEL assay after 24 h treatment, and the destabilization of lysosome and mitochondria were early upstream initiating events. This study showed that cathepsin D played a crucial role in the process of apoptosis. The release of cathepsin D to the cytosol appeared to be an early event that preceded the release of cytochrome c from mitochondria. Moreover, inhibition of cathepsin D activity resulted in suppressed release of cytochrome c. To further determine the involvement of oxidative stress in 6‐gingerol‐induced apoptosis, the intracellular generation of reactive oxygen species (ROS) and reduced glutathione (GSH) were examined. Taken together, these results suggest that cathepsin D may be a positive mediator of 6‐gingerol induced apoptosis in HepG2 cells, acting upstream of cytochrome c release, and the apoptosis may be associated with oxidative stress. Copyright


Neurotoxicology | 2012

Arsenic-induced inhibition of hippocampal neurogenesis and its reversibility

Shuang Liu; Fengyuan Piao; Xiance Sun; Lulu Bai; Yan Peng; Yuanxia Zhong; Ning Ma; Wenchang Sun

Arsenic exposure can result in damages of the neurological system. The present study aimed at whether cell proliferation and neurogenesis in the adult mouse hippocampus were affected after arsenic exposure and whether they could recover after exposure cessation. Mice were randomly placed into 3 groups. The first group received distilled water alone for 4 months (control group); the second group received 4.0 mg/L As(2)O(3) through drinking water for 4 months (arsenic group); the third group received 4.0 mg/L As(2)O(3) for 2 months and then changed to distilled water for another 2 months (recovery group). Serum and cerebrum arsenic concentrations of the arsenic group were significantly elevated, and then decreased to normal after the change of arsenic to water in the diet. After a four-month administration, the hippocampal number of proliferative cells and the percentage of new mature neurons decreased in the arsenic group as compared with the control group, however, increased significantly in the recovery group when compared with the arsenic group, and restored to the control level. There were no significant differences for apoptosis in different groups. Obvious histopathological ameliorations were observed in the hippocampus of the recovery group. The inhibition of hippocampus cell proliferation and neurogenesis by arsenic is reversible after the arsenic administration was terminated.


Food and Chemical Toxicology | 2014

Perfluorooctane sulfonate blocked autophagy flux and induced lysosome membrane permeabilization in HepG2 cells.

Xiaofeng Yao; Jun Cao; Liming Xu; Xiance Sun; Jian Kang; Guang Yang; Liping Jiang; Chengyan Geng; Chuan-Zhou Gao; Laifu Zhong; Yufang Ma

Perfluorooctane sulfonate (PFOS) is an emerging persistent organic pollutant widely distributed in the environment, wildlife and human. In this study, as observed under the transmission electron microscope, PFOS increased autophagosome numbers in HepG2 cells, and it was confirmed by elevated LC3-II levels in Western blot analysis. PFOS increased P62 level and chloroquine failed to further increase the expression of LC3-II after PFOS treatment, indicating that the accumulation of autophagosome was due to impaired degradation rather than increased formation. With acridine orange staining, we found PFOS caused lysosomal membrane permeabilization (LMP). In this study, autophasome formation inhibitor 3-methyladenine protected cells against PFOS toxicity, autophagy stimulator rapamycin further decreased cell viability and increased LDH release, cathepsin inhibitor did not influence cell viability of PFOS-treated HepG2 cells significantly. These further supported the notion that autophagic cell death contributed to PFOS-induced hepatotoxicity. In summary, the data of the present study revealed that PFOS induced LMP and consequent blockage of autophagy flux, leading to an excessive accumulation of the autophagosomes and turning autophagy into a destructive process eventually. This finding will provide clues for effective prevention and treatment of PFOS-induced hepatic disease.


Food and Chemical Toxicology | 2014

Sodium arsenite induces ROS-dependent autophagic cell death in pancreatic β-cells

Xue-Xin Zhu; Xiaofeng Yao; Liping Jiang; Chengyan Geng; Laifu Zhong; Guang Yang; Bai-Lu Zheng; Xiance Sun

Inorganic arsenic is a worldwide environmental pollutant. Inorganic arsenics positive relationship with the incidence of type 2 diabetes mellitus arouses concerns associated with its etiology in diabetes among the general human population. In this study, the inhibitor of autophagosome formation, 3-methyladenine, protected the cells against sodium arsenite cytotoxicity, and the autophagy stimulator rapamycin further decreased the cell viability of sodium arsenite-treated INS-1 cells. These finding suggested the hypothesis that autophagic cell death contributed to sodium arsenite-induced cytotoxicity in INS-1 cells. Sodium arsenite increased the autophagosome-positive puncta in INS-1 cells observed under a fluorescence microscope, and this effect was confirmed by the elevated LC3-II levels detected through Western blot. The LC3 turnover assay indicated that the accumulation of autophagosomes in the arsenite-treated INS-1 cells was due to increased formation rather than impaired degradation. The pretreatment of INS-1 cells with the ROS inhibitor NAC reduced autophagosome formation and reversed the sodium arsenite cytotoxicity, indicating that sodium arsenite-induced autophagic cell death was ROS-dependent. In summary, the precise molecular mechanisms through which arsenic is related to diabetes have not been completely elucidated, but the ROS-dependent autophagic cell death of pancreatic β-cells described in this study may help to elucidate the underlying mechanism.


Toxicon | 2015

Citreoviridin induces ROS-dependent autophagic cell death in human liver HepG2 cells.

Yanan Liu; Yuexia Wang; Xiaofang Liu; Liping Jiang; Guang Yang; Xiance Sun; Chengyan Geng; Qiujuan Li; Min Chen; Xiaofeng Yao

Citreoviridin (CIT) is one of toxic mycotoxins derived from fungal species in moldy cereals. Whether CIT exerts hepatotoxicity and the precise molecular mechanisms of CIT hepatotoxicity are not completely elucidated. In this study, the inhibitor of autophagosome formation, 3-methyladenine, protected the cells against CIT cytotoxicity, and the autophagy stimulator rapamycin further decreased the cell viability of CIT-treated HepG2 cells. Knockdown of Atg5 with Atg5 siRNA alleviated CIT-induced cell death. These finding suggested the hypothesis that autophagic cell death contributed to CIT-induced cytotoxicity in HepG2 cells. CIT increased the autophagosome number in HepG2 cells observed under a transmission electron microscope, and this effect was confirmed by the elevated LC3-II levels detected through Western blot. Reduction of P62 protein levels and the result of LC3 turnover assay indicated that the accumulation of autophagosomes in the CIT-treated HepG2 cells was due to increased formation rather than impaired degradation. The pretreatment of HepG2 cells with the ROS inhibitor NAC reduced autophagosome formation and reversed the CIT cytotoxicity, indicating that CIT-induced autophagic cell death was ROS-dependent. In summary, ROS-dependent autophagic cell death of HpeG2 cells described in this study may help to elucidate the underlying mechanism of CIT cytotoxicity.


Toxins | 2015

Citreoviridin Induces Autophagy-Dependent Apoptosis through Lysosomal-Mitochondrial Axis in Human Liver HepG2 Cells.

Yuexia Wang; Yanan Liu; Xiaofang Liu; Liping Jiang; Guang Yang; Xiance Sun; Chengyan Geng; Qiujuan Li; Xiaofeng Yao; Min Chen

Citreoviridin (CIT) is a mycotoxin derived from fungal species in moldy cereals. In our previous study, we reported that CIT stimulated autophagosome formation in human liver HepG2 cells. Here, we aimed to explore the relationship of autophagy with lysosomal membrane permeabilization and apoptosis in CIT-treated cells. Our data showed that CIT increased the expression of LC3-II, an autophagosome biomarker, from the early stage of treatment (6 h). After treatment with CIT for 12 h, lysosomal membrane permeabilization occurred, followed by the release of cathepsin D in HepG2 cells. Inhibition of autophagosome formation with siRNA against Atg5 attenuated CIT-induced lysosomal membrane permeabilization. In addition, CIT induced collapse of mitochondrial transmembrane potential as assessed by JC-1 staining. Furthermore, caspase-3 activity assay showed that CIT induced apoptosis in HepG2 cells. Inhibition of autophagosome formation attenuated CIT-induced apoptosis, indicating that CIT-induced apoptosis was autophagy-dependent. Cathepsin D inhibitor, pepstatin A, relieved CIT-induced apoptosis as well, suggesting the involvement of the lysosomal-mitochondrial axis in CIT-induced apoptosis. Taken together, our data demonstrated that CIT induced autophagy-dependent apoptosis through the lysosomal-mitochondrial axis in HepG2 cells. The study thus provides essential mechanistic insight, and suggests clues for the effective management and treatment of CIT-related diseases.


Scientific Reports | 2016

Taurine protects against As2O3-induced autophagy in livers of rat offsprings through PPARγ pathway.

Jie Bai; Xiaofeng Yao; Liping Jiang; Qiaoting Zhang; Huai Guan; Shuang Liu; Wei Wu; Tianming Qiu; Ni Gao; Lei Yang; Guang Yang; Xiance Sun

Chronic exposures to arsenic had been associated with metabolism diseases. Peroxisome proliferator-activated receptor gamma (PPARγ) was found in the liver, regulated metabolism. Here, we found that the expression of PPARγ was decreased, the generation of reactive oxygen species (ROS) and autophagy were increased after treatment with As2O3 in offsprings’ livers. Taurine (Tau), a sulfur-containing β–amino acid could reverse As2O3-inhibited PPARγ. Tau also inhibit the generation of ROS and autophagy. We also found that As2O3 caused autophagic cell death and ROS accelerated in HepG2 cells. Before incubation with As2O3, the cells were pretreated with PPARγ activator Rosiglitazone (RGS), we found that autophagy and ROS was inhibited in HepG2 cells, suggesting that inhibition of PPARγ contributed to As2O3-induced autophagy and the generation of ROS. After pretreatment with Tau, the level of PPARγ was improved and the autophagy and ROS was inhibited in As2O3-treated cells, suggesting that Tau could protect hepatocytes against As2O3 through modulating PPARγ pathway.


Chemico-Biological Interactions | 2016

6-Gingerol induces autophagy to protect HUVECs survival from apoptosis

Shaopeng Wang; Xiance Sun; Liping Jiang; Xiaofang Liu; Min Chen; Xiaofeng Yao; Qinghua Sun; Guang Yang

6-Gingerol, the major pharmacologically-active component of ginger, has the potential to prevent heart disease. However, the mechanisms are not well understood. In this study, the protective effect of 6-gingerol against hydrogen peroxide-induced apoptosis in human umbilical vein endothelial cells (HUVECs) was investigated. Apoptosis was detected by Hoechst 33342 and Flow cytometry analysis. To further elucidate the crosstalk between apoptosis and autophagy, we tested the expression of autophagy related proteins, LC3B, Bcl-2, Beclin1, AKT, p-AKT, mechanistic target of rapamycin (mTOR), and p-mTOR. Furthermore, mitochondrial membrane potential and the intracellular generation of reactive oxygen species (ROS) were also investigated. Our data revealed that 6-gingerol significantly reduced apoptosis by inducing autophagy. It has been demonstrated that 6-gingerol suppressed the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR signaling pathway, increased the expression of Beclin1 to promote autophagy, and increased Bcl-2 expression to inhibit apoptosis. In addition, the damage of mitochondrial was protected, and ROS level was decreased by 6-gingerol. These firmly indicate 6-gingerol has a strong protective ability against the apoptosis caused by oxidative stress in HUVECs, and the mechanism may relate to the induction of autophagy. Our data suggest 6-gingerol may be beneficial in the prevention of atherosclerosis.

Collaboration


Dive into the Xiance Sun's collaboration.

Top Co-Authors

Avatar

Liping Jiang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Guang Yang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaofeng Yao

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaofang Liu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Chengyan Geng

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Min Chen

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Shuang Liu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Laifu Zhong

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Fengyuan Piao

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Shaopeng Wang

Dalian Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge