Network


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

Hotspot


Dive into the research topics where Xiaoyan Yuan is active.

Publication


Featured researches published by Xiaoyan Yuan.


Oncotarget | 2016

Repeated PM2.5 exposure inhibits BEAS-2B cell P53 expression through ROS-Akt-DNMT3B pathway-mediated promoter hypermethylation

Wei Zhou; Dongdong Tian; Jun He; Yi-Mei Wang; Lijun Zhang; Lan Cui; Li Jia; Li Zhang; Lizhong Li; Yulei Shu; Shouzhong Yu; Jun Zhao; Xiaoyan Yuan; Shuangqing Peng

Long-term exposure to fine particulate matter (PM2.5) has been reported to be closely associated with the increased lung cancer risk in populations, but the mechanisms underlying PM-associated carcinogenesis are not yet clear. Previous studies have indicated that aberrant epigenetic alterations, such as genome-wide DNA hypomethylation and gene-specific DNA hypermethylation contribute to lung carcinogenesis. And silence or mutation of P53 tumor suppressor gene is the most prevalent oncogenic driver in lung cancer development. To explore the effects of PM2.5 on global and P53 promoter methylation changes and the mechanisms involved, we exposed human bronchial epithelial cells (BEAS-2B) to low concentrations of PM2.5 for 10 days. Our results indicated that PM2.5-induced global DNA hypomethylation was accompanied by reduced DNMT1 expression. PM2.5 also induced hypermethylation of P53 promoter and inhibited its expression by increasing DNMT3B protein level. Furthermore, ROS-induced activation of Akt was involved in PM2.5-induced increase in DNMT3B. In conclusion, our results strongly suggest that repeated exposure to PM2.5 induces epigenetic silencing of P53 through ROS-Akt-DNMT3B pathway-mediated promoter hypermethylation, which not only provides a possible explanation for PM-induced lung cancer, but also may help to identify specific interventions to prevent PM-induced lung carcinogenesis.


Journal of Environmental Sciences-china | 2014

T-2 toxin induces developmental toxicity and apoptosis in zebrafish embryos

Guogang Yuan; Yimei Wang; Xiaoyan Yuan; Tingfen Zhang; Jun Zhao; Liuyu Huang; Shuangqing Peng

T-2 toxin is one of the most important trichothecene mycotoxins occurring in various agriculture products. The developmental toxicity of T-2 toxin and the exact mechanism of action at early life stages are not understood precisely. Zebrafish embryos were exposed to different concentrations of the toxin at 4-6 hours post fertilization (hpf) stage of development, and were observed for different developmental toxic effects at 24, 48, 72, and 144 hpf. Exposure to 0.20 μmol/L or higher concentrations of T-2 toxin significantly increased the mortality and malformation rate such as tail deformities, cardiovascular defects and behavioral changes in early developmental stages of zebrafish. T-2 toxin exposure resulted in significant increases in reactive oxygen species (ROS) production and cell apoptosis, mainly in the tail areas, as revealed by Acridine Orange staining at 24 hpf. In addition, T-2 toxin-induced severe tail deformities could be attenuated by co-exposure to reduced glutathione (GSH). T-2 toxin and GSH co-exposure induced a significant decrease of ROS production in the embryos. The overall results demonstrate that T-2 toxin is able to produce oxidative stress and induce apoptosis, which are involved in the developmental toxicity of T-2 toxin in zebrafish embryos.


Ecotoxicology and Environmental Safety | 2015

Combined exposure to nano-silica and lead induced potentiation of oxidative stress and DNA damage in human lung epithelial cells.

Chun-Feng Lu; Xiaoyan Yuan; Lizhong Li; Wei Zhou; Jun Zhao; Yi-Mei Wang; Shuangqing Peng

Growing evidence has confirmed that exposure to ambient particulate matters (PM) is associated with increased morbidity and mortality of cardiovascular and pulmonary diseases. Ambient PM is a complex mixture of particles and air pollutants. Harmful effects of PM are specifically associated with ultrafine particles (UFPs) that can adsorb high concentrations of toxic air pollutants and are easily inhaled into the lungs. However, combined effects of UFPs and air pollutants on human health remain unclear. In the present study, we elucidated the combined toxicity of silica nanoparticles (nano-SiO2), a typical UFP, and lead acetate (Pb), a typical air pollutant. Lung adenocarcinoma A549 cells were exposed to nano-SiO2 and Pb alone or their combination, and their combined toxicity was investigated by focusing on cellular oxidative stress and DNA damage. Factorial analyses were performed to determine the potential interactions between nano-SiO2 and Pb. Our results showed that exposure of A549 cells to a modest cytotoxic concentration of Pb alone induced oxidative stress, as evidenced by elevated reactive oxygen species generation and lipid peroxidation, and reduced glutathione content and superoxide dismutase and glutathione peroxidase activities. In addition, exposure of A549 cells to Pb alone induced DNA damage, as evaluated by alkaline comet assay. Exposure of A549 cells to non-cytotoxic concentration of nano-SiO2 did not induce cellular oxidative stress and DNA damage. However, exposure to the combination of nano-SiO2 and Pb potentiated oxidative stress and DNA damage in A549 cells. Factorial analyses indicated that the potentiation of combined toxicity of nano-SiO2 and Pb was induced by additive or synergistic interactions.


Autophagy | 2016

TP53-dependent autophagy links the ATR-CHEK1 axis activation to proinflammatory VEGFA production in human bronchial epithelial cells exposed to fine particulate matter (PM2.5)

Xiuduan Xu; Hongli Wang; Shasha Liu; Chen Xing; Yang Liu; Aodengqimuge; Wei Zhou; Xiaoyan Yuan; Yongfu Ma; Meiru Hu; Yongliang Hu; Shuxian Zou; Ye Gu; Shuangqing Peng; Shengtao Yuan; Weiping Li; Yuanfang Ma; Lun Song

ABSTARCT Epidemiological and clinical studies have increasingly shown that fine particulate matter (PM2.5) is associated with a number of pathological respiratory diseases, such as bronchitis, asthma, and chronic obstructive pulmonary disease, which share the common feature of airway inflammation induced by particle exposure. Thus, understanding how PM2.5 triggers inflammatory responses in the respiratory system is crucial for the study of PM2.5 toxicity. In the current study, we found that exposing human bronchial epithelial cells (immortalized Beas-2B cells and primary cells) to PM2.5 collected in the winter in Wuhan, a city in southern China, induced a significant upregulation of VEGFA (vascular endothelial growth factor A) production, a signaling event that typically functions to control chronic airway inflammation and vascular remodeling. Further investigations showed that macroautophagy/autophagy was induced upon PM2.5 exposure and then mediated VEGFA upregulation by activating the SRC (SRC proto-oncogene, non-receptor tyrosine kinase)-STAT3 (signal transducer and activator of transcription 3) pathway in bronchial epithelial cells. By exploring the upstream signaling events responsible for autophagy induction, we revealed a requirement for TP53 (tumor protein p53) activation and the expression of its downstream target DRAM1 (DNA damage regulated autophagy modulator 1) for the induction of autophagy. These results thus extend the role of TP53-DRAM1-dependent autophagy beyond cell fate determination under genotoxic stress and to the control of proinflammatory cytokine production. Moreover, PM2.5 exposure strongly induced the activation of the ATR (ATR serine/threonine kinase)-CHEK1/CHK1 (checkpoint kinase 1) axis, which subsequently triggered TP53-dependent autophagy and VEGFA production in Beas-2B cells. Therefore, these findings suggest a novel link between processes regulating genomic integrity and airway inflammation via autophagy induction in bronchial epithelial cells under PM2.5 exposure.


Environmental Toxicology | 2016

PM2.5 induces embryonic growth retardation: Potential involvement of ROS‐MAPKs‐apoptosis and G0/G1 arrest pathways

Xiaoyan Yuan; Yi-Mei Wang; Lizhong Li; Wei Zhou; Dongdong Tian; Chunfeng Lu; Shouzhong Yu; Jun Zhao; Shuangqing Peng

Airborne fine particulate matter (PM2.5) is an “invisible killer” to human health. There is increasing evidence revealing the adverse effects of PM2.5 on the early embryonic development and pregnancy outcome, but the molecular mechanism underlying PM2.5‐induced embryotoxicity is largely unknown. Previous studies have documented that exposure to PM triggers ROS generation, leads to subsequent activation of MAPKs signaling, and results in corresponding cell biological changes including enhanced apoptosis and altered cell cycle in the cardiopulmonary system. Here, we investigated whether ROS‐MAPKs‐apoptosis/cell cycle arrest pathways play an important role in PM2.5‐induced embryotoxicity using the rat whole embryo culture system. The results showed that PM2.5 treatment led to embryonic growth retardation at concentrations of 50 μg/ml and above, as evidenced by the reduced yolk sac diameter, crown‐rump length, head length and somite number. PM2.5‐induced embryonic growth retardation was accompanied by cell apoptosis and G0/G1 phase arrest. Furthermore, ROS generation and subsequent activation of JNK and ERK might be involved in PM2.5‐induced apoptosis and G0/G1 phase arrest by downregulating Bcl‐2/Bax protein ratio and upregulating p15INK4B, p16INK4A, and p21WAF1/CIP1 transcription level. In conclusion, our results indicate that ROS‐JNK/ERK‐apoptosis and G0/G1 arrest pathways are involved in PM2.5‐induced embryotoxicity, which not only provides insights into the molecular mechanism of PM2.5‐induced embryotoxicity, but also may help to identify specific interventions to improve adverse pregnancy outcomes of PM2.5.


Toxicology Letters | 2014

SUMO-specific protease 1 modulates cadmium-augmented transcriptional activity of androgen receptor (AR) by reversing AR SUMOylation

Yaxiong Cui; Xiaoyan Yuan; Haitao Yuan; Yi-Mei Wang; Jun He; Jun Zhao; Shuangqing Peng

Cadmium is a potential prostate carcinogen and can mimic the effects of androgen by a mechanism that involves the hormone-binding domain of the androgen receptor (AR), which is a key transcriptional factor in prostate carcinogenesis. We focused on transcriptional activity of AR to investigate the toxicity of cadmium exposure on human prostate cell lines. Cadmium increased the proliferative index of LNCaP and the proliferative effect was obstructed significantly by AR blocking agent. In luciferase assay, cadmium activated the transcriptional activity of AR in 293T cells co-transfected with wild-type AR and an ARE (AR response elements)-luciferase reporter gene. Cadmium also increased expression of PSA, a downstream gene of AR, whereas the metal had no significant effect on AR amount. AR is regulated by multiple posttranslational modifications including SUMOylation. SUMOylated AR shows a lower transcriptional activity. SUMO-specific protease 1 (SENP1) decreases AR SUMOylation by deconjugating AR-SUMO covalent bond. We detected that cadmium increased the amount of SENP1 in a dose and time dependent manner. Knocking down of SENP1 by RNAi led to decrease of PSA expression and transcriptional activity of AR in luciferase assay. Furthermore, co-immunoprecipitation (Co-IP) results showed that SUMOylation level of AR was decreased after cadmium treatment. In conclusion, our results indicated that cadmium-induced SENP1 enhanced AR transcriptional activity by decreasing AR SUMOylation.


Ecotoxicology and Environmental Safety | 2017

Overexpression of HO-1 assisted PM2.5-induced apoptosis failure and autophagy-related cell necrosis

Wei Zhou; Xiaoyan Yuan; Li Zhang; Baoting Su; Dongdong Tian; Yang Li; Jun Zhao; Yi-Mei Wang; Shuangqing Peng

Severe smog/haze events accompanied by extremely high concentrations of airborne fine particulate matter (PM2.5) have emerged frequently in China and the potential health risks have attracted ever-growing attention. During these episodes, a surge in hospital visits for acute respiratory symptoms and respiratory diseases exacerbation has been reported to be associated with acute exposure to high-levels of particulate matters. To investigate cell fate determination and the underlying pathogenic mechanisms during severe haze episodes or smog events, we exposed human lung epithelial cells (BEAS-2B) to PM2.5 (0-400μg/mL) for 24h and found that high doses of PM2.5 caused cell necrosis and autophagy dysfunction, while co-treatment with the autophagy inhibitor 3-MA could partially reduce PM2.5-induced cell necrosis. Exposure to PM2.5 also increased the expression and mitochondrial transposition of heme oxygenase 1 (HO-1), which consequently reduced the release of cytochrome C from mitochondria to cytosol. Knockdown of HO-1 by siRNA attenuated the mitochondrial accumulation of HO-1, reversed HO-1-induced the reduction of cytochrome C release and promoted PM2.5-induced cell apoptosis. In contrast to necrosis, PM2.5-induced autophagy was independent of HO-1. In conclusion, our results demonstrate that acute exposure to high PM2.5 concentrations causes autophagy-related cell necrosis. The decrease in cytochrome C release and apoptosis by upregulation of HO-1 maybe assist PM2.5-induced autophagy-related cell necrosis. Further, this study reveals dual roles for HO-1 in PM2.5-induced cytotoxicity and presents a possible explanation for the onset of acute respiratory symptoms under extreme particulate air pollution.


Regulatory Toxicology and Pharmacology | 2017

Subchronic toxicity study of yttrium nitrate by 90-day repeated oral exposure in rats

Yi-Mei Wang; Zhou Yu; Zeng-Ming Zhao; Li Jia; Haiqin Fang; Tingfen Zhang; Xiaoyan Yuan; Yulei Shu; Jun He; Hui Peng; Lizhong Li; Jun Zhao; Xudong Jia; Shuangqing Peng

ABSTRACT Concerns regarding the adverse effects of long‐term exposure to low levels of rare earth elements (REEs) from foods on human health have arisen in recent years. Nevertheless, no official acceptable daily intake (ADI) has yet been proposed for either total REEs or individual REE. In accordance with the Organization for Economic Co‐operation and Development (OECD) testing guideline, the present study was undertaken to evaluate the subchronic toxicity of yttrium, a representative heavy REE with higher contaminated level in foods in China, to achieve a no observed adverse effect level (NOAEL) which is a critical basis for the establishment of an ADI. Yttrium nitrate was orally administered to rats at doses of 0, 10, 30 and 90 mg/kg/day for 90 days followed by a recovery period of 4 weeks. The following toxicity indices were measured: mortality, clinical signs, daily food consumption and weekly body weight; urinalysis, hematology, blood coagulation, clinical biochemistry and histopathology at the end of administration and recovery periods. No toxicologically significant changes were found in any yttrium‐treated group as compared to the concurrent control group. Under the present experimental condition, the NOAEL in rats was thus set at 90 mg/kg for yttrium nitrate, i.e. 29.1 mg/kg for yttrium. HighlightsSubchronic toxicity study was performed in accordance with OECD TG 408.The NOAEL in rats was estimated to be 29.1 mg/kg for yttrium.Daily intake of yttrium from foods is acceptable for adults in China.


Frontiers in Pharmacology | 2017

Impairment of Mitochondrial Biogenesis and Dynamics Involved in Isoniazid-Induced Apoptosis of HepG2 Cells Was Alleviated by p38 MAPK Pathway

Tian-Guang Zhang; Takashi Ikejima; Lizhong Li; Xiaoyan Yuan; Jun Zhao; Yi-Mei Wang; Shuangqing Peng

Isoniazid (INH), a widely used first-line antitubercular drug, has been noted to be associated with hepatotoxicity. In spite of extensive researches over many decades, the mechanism of INH-induced hepatotoxicity still remains poorly understood. Recently, mitochondrial toxicity has been emerging as a new paradigm for INH-induced hepatotoxicity. In this study, we showed that INH impaired mitochondrial biogenesis and dynamics in human hepatocarcinoma HepG2 cells. INH reduced mitochondrial membrane potential (MMP) and induced mitochondria swelling. INH also inhibited the protein expressions of three major mitochondrial biogenesis regulators, SIRT1, PGC1α and NRF1, along with increased acetylation of PGC1α. Meanwhile, INH decreased the number of mitochondria, accompanied by decreased expression of mitochondrial protein COX IV. INH caused mitochondrial fragmentation involving decreased levels of the fusion protein MFN2 as well as the fission protein DRP1. INH-reduced DRP1 expression was associated with the increase of apoptosis, suggesting the existence of pro-survival fission and its involvement in mitochondrial quality control. INH activated p38 MAPK, whereas inhibition of p38 MAPK aggravated INH-induced decreases of SIRT1, PGC1α, NRF1, COX IV and DRP1 expressions. P38 MAPK inhibition also further up-regulated the acetylation of PGC1α and exacerbated INH-induced MMP loss, mitochondrial swelling and apoptosis. Taken together, INH-activated p38 MAPK induced mitochondrial biogenesis to alleviate apoptosis through partly recovering SIRT1-PGC1α pathway activation. In the meantime, p38 MAPK activation by INH promoted protective mitochondrial fission to alleviate apoptosis by partial recovery of DRP1 expression.


Journal of Applied Toxicology | 2018

Prolonged exposure to carbon nanoparticles induced methylome remodeling and gene expression in zebrafish heart: CNP exposure caused cardiac inflammation via methylome remodeling

Wei Zhou; Dongdong Tian; Jun He; Xiabei Yan; Jun Zhao; Xiaoyan Yuan; Shuangqing Peng

Growing black carbon (BC) emission has become one of the major urgent environmental issues facing human beings. Usually, BC or BC‐containing carbon nanoparticles (CNPs) were recognized as non‐directly toxic components of atmospheric particulate matter. However, epidemiology studies have provided much evidence of the associations of exposure of particulate‐containing carbon particles with cardiovascular diseases. There are still no related studies to support the epidemiological conclusions. Hence, in this article we exposed adult zebrafish to CNPs for 60 days, and then explored the heart location and potential adverse effects on cardiac tissues of these nanosized carbon particles. Our results first showed direct visualization of cardiac endothelial uptake and heart deposition of CNPs in zebrafish. In addition, CNPs caused significant ultrastructural alterations in myocardial tissue and induced the expression of inflammatory cytokines in a dose‐dependent manner, resulting in sub‐endocardial inflammation and cell apoptosis. Moreover, our data demonstrated the perturbations caused by CNPs on DNA methylation, suggesting that DNA methylome remodeling might play a critical role in CNP‐induced cardiotoxicity in zebrafish heart. Therefore, this study not only proved a laboratory link between CNP exposure and cardiotoxicity in vivo, but also indicated a possible toxicity mechanism involved.

Collaboration


Dive into the Xiaoyan Yuan's collaboration.

Top Co-Authors

Avatar

Shuangqing Peng

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jun Zhao

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Yi-Mei Wang

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lizhong Li

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Dongdong Tian

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jun He

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Li Zhang

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Li Jia

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Lijun Zhang

Academy of Military Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge