Network


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

Hotspot


Dive into the research topics where Yanfeng Shi is active.

Publication


Featured researches published by Yanfeng Shi.


Science of The Total Environment | 2017

1H NMR-based metabolomics study on repeat dose toxicity of fine particulate matter in rats after intratracheal instillation

Yannan Zhang; Hejing Hu; Yanfeng Shi; Xiaozhe Yang; Lige Cao; Jing Wu; Collins Otieno Asweto; Lin Feng; Junchao Duan; Zhiwei Sun

Systemic metabolic effects and toxicity mechanisms of ambient fine particulate matter (PM2.5) remain uncertain. In order to investigate the mechanisms in PM2.5 toxicity, we explored the endogenous metabolic changes and possible influenced metabolic pathways in rats after intratracheal instillation of PM2.5 by using a 1H nuclear magnetic resonance (NMR)-based metabolomics approach. Liver and kidney histopathology examinations were also performed. Chemical characterization demonstrated that PM2.5 was a complex mixture of elements. Histopathology showed cellular edema in liver and glomerulus atrophy of the PM2.5 treated rats. We systematically analyzed the metabolites changes of serum and urine in rats using 1H NMR techniques in combination with multivariate statistical analysis. Significantly reduced levels of lactate, alanine, dimethylglycine, creatine, glycine and histidine in serum, together with increased levels of citrate, arginine, hippurate, allantoin and decreased levels of allthreonine, lactate, alanine, acetate, succinate, trimethylamine, formate in urine were observed of PM2.5 treated rats. The mainly affected metabolic pathways by PM2.5 were glycine, serine and threonine metabolism, glyoxylate and dicarboxylate metabolism, citrate cycle (TCA cycle), nitrogen metabolism and methane metabolism. Our study provided important information on assessing the toxicity of PM2.5 and demonstrated that metabolomics approach can be employed as a tool to understand the toxicity mechanism of complicated environmental pollutants.


Chemosphere | 2017

Multi-organ toxicity induced by fine particulate matter PM2.5 in zebrafish (Danio rerio) model

Junchao Duan; Hejing Hu; Yannan Zhang; Lin Feng; Yanfeng Shi; Mark R. Miller; Zhiwei Sun

The fine particulate matter (PM2.5) in air pollution is a major public health concern and now known to contribute to severe diseases, therefore, a comprehensive understanding of PM2.5-induced adverse effects in living organisms is needed urgently. This study was aimed to evaluate the toxicity of PM2.5 on multi-organ systems in a zebrafish (Danio rerio) model. The embryonic toxicity induced by PM2.5 was demonstrated by an increase in mortality and inhibition of hatching rate, in a dose- and time-dependent manner. PM2.5 caused the pericardial edema, as well as reducing heart rate and cardiac output. The area of sub-intestinal vessels (SIVs) was significant reduced in Tg(fli-1:EGFP) transgenic zebrafish lines. Morphological defects and yolk sac retention were associated with hepatocyte injury. In addition, PM2.5 disrupted the axonal integrity, altering of axon length and pattern in Tg(NBT:EGFP) transgenic lines. Genes involved in cardiac function (spaw, supt6h, cmlc1), angiogenesis (vegfr2a, vegfr2b), and neural function (gabrd, chrna3, npy8br) were markedly down-regulated; while genes linked to hepatic metabolism (cyp1a, cyp1b1, cyp1c1) were significantly up-regulated by PM2.5. In summary, our data showed that PM2.5 induced the cardiovascular toxicity, hepatotoxicity and neurotoxicity in zebrafish, suggested that PM2.5 could cause multi-organ toxicity in aquatic organism.


Environmental Science and Pollution Research | 2017

Genome-wide transcriptional analysis of cardiovascular-related genes and pathways induced by PM2.5 in human myocardial cells

Lin Feng; Xiaozhe Yang; Collins Otieno Asweto; Jing Wu; Yannan Zhang; Hejing Hu; Yanfeng Shi; Junchao Duan; Zhiwei Sun

Air pollution has been a major environment-related health threat. Most of the studies on PM2.5 toxicity have verified on the cardiovascular system and endothelial cells. However, researches on PM2.5-induced myocardial-related toxicity are limited. This study aims to fully understand the toxic effects of PM2.5 on human myocardial cell (AC16) and explore its molecular mechanism based on microarray analysis and bioinformatics analysis. Microarray data analysis manifested that PM2.5-induced toxicity affected expression of 472 genes compared with the control group, including 166 upregulated genes and 306 downregulated genes in human myocardial (AC16) cells. GO analysis showed that cellular processes such as immune response, cell maturation, embryonic heart tube morphogenesis, cellular response to electrical stimulus, skeletal muscle tissue regeneration, and negative regulation of signal transduction were upregulated, while regulation of transcription (DNA-dependent), rhythmic process, protein destabilization apoptotic process, and innate immune response were downregulated. The pathway analysis indicates that cell signaling pathways such as cytokine-cytokine receptor interaction, NF-κB signaling pathway, chemokine signaling pathway, endocrine and other factor-regulated calcium reabsorption, HTLV-I infection, and cell adhesion molecules (CAMs) were upregulated, while the TGF-β signaling pathway was downregulated. In addition, Signal-net showed that the TUBA4A, ADRBK2, BRIX1, SMC4, EIF5B, PRMT1, ATG4B, and NDC80 genes were significantly decreased, while the expression of the KRT6B gene was markedly increased compared with the control group. All the genes were verified by qRT-PCR. This study had provided new bioinformatics evidences in PM2.5-induced myocardial tissue toxicity which is necessary for further cardiovascular system toxicity studies.


Ecotoxicology and Environmental Safety | 2018

Cytotoxicity induced by fine particulate matter (PM 2.5 ) via mitochondria-mediated apoptosis pathway in human cardiomyocytes

Xiaozhe Yang; Lin Feng; Yannan Zhang; Hejing Hu; Yanfeng Shi; Shuang Liang; Tong Zhao; Yang Fu; Junchao Duan; Zhiwei Sun

Although the strongly causal associations were between fine particulate matter (PM2.5) and cardiovascular disease, the toxic effect and potential mechanism of PM2.5 on heart was poorly understood. Thus, the aim of this study was to evaluate the cardiac toxicity of PM2.5 exposure on human cardiomyocytes (AC16). The cell viability was decreased while the LDH release was increased in a dose-dependent way after AC16 exposed to PM2.5. The reactive oxygen species (ROS) generation and production of malondialdehyde (MDA) were increased followed by the decreasing in superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). The damage of mitochondria was observed by ultra-structural analysis and MMP measurement. The apoptotic rate of AC16 were markedly elevated which was triggered by PM2.5. In addition, the proteins involved in mitochondria- mediated apoptosis pathway were measured. The protein levels of Caspase-3, Caspase-9 and Bax were up-regulated while the anti-apoptotic protein, Bcl-2 was down-regulated after AC16 exposed to PM2.5. In summary, our results demonstrated that mitochondria-mediated apoptosis pathway played a critical role in PM2.5-induced myocardial cytotoxicity in AC16, which suggested that PM2.5 may contribute to cardiac dysfunction.


Environmental Science and Pollution Research | 2017

Low-dose combined exposure of nanoparticles and heavy metal compared with PM2.5 in human myocardial AC16 cells

Lin Feng; Xiaozhe Yang; Collins Otieno Asweto; Jing Wu; Yannan Zhang; Hejing Hu; Yanfeng Shi; Junchao Duan; Zhiwei Sun

The co-exposure toxicity mechanism of ultrafine particles and pollutants on human cardiovascular system are still unclear. In this study, the combined effects of silica nanoparticles (SiNPs) and/or carbon black nanoparticles (CBNPs) with Pb(AC)2 compared with particulate matter (PM)2.5 were investigated in human myocardial cells (AC16). Our study detected three different combinations of SiNPs and Pb(AC)2, CBNPs and Pb(AC)2, and SiNPs and CBNPs compared with PM2.5 at low-dose exposure. Using PM2.5 as positive control, our results suggested that the combination of SiNPs and Pb(AC)2/CBNPs could increase the production of reactive oxygen species (ROS), lactate dehydrogenase leakage (LDH), and malondialdehyde (MDA) and decrease the activities of superoxide dismutase (SOD) and glutathione (GSH); induce inflammation by the upregulation of protein CRP and TNF-α, and apoptosis by the upregulation of protein caspase-3, caspase-9, and Bax while the downregulation of protein Bcl-2; and trigger G2/M phase arrest by the upregulation of protein Chk2 and downregulation of protein Cdc2 and cyclin B1. In addition, the combination of CBNPs and Pb(AC)2 induced a significant increase in MDA and reduced the activities of ROS, LDH, SOD, and GSH, with G1/S phase arrest via upregulation of Chk1 and downregulation of CDK6 and cyclin D1. Our data suggested that the additive interaction and synergistic interaction are the major interaction in co-exposure system, and PM2.5 could trigger more severe oxidative stress, G2/M arrest, and apoptosis than either co-exposure or single exposure.


Chemosphere | 2017

Microarray-based bioinformatics analysis of the combined effects of SiNPs and PbAc on cardiovascular system in zebrafish

Hejing Hu; Yannan Zhang; Yanfeng Shi; Lin Feng; Junchao Duan; Zhiwei Sun

With rapid development of nanotechnology and growing environmental pollution, the combined toxic effects of SiNPs and pollutants of heavy metals like lead have received global attentions. The aim of this study was to explore the cardiovascular effects of the co-exposure of SiNPs and lead acetate (PbAc) in zebrafish using microarray and bioinformatics analysis. Although there was no other obvious cardiovascular malformation except bleeding phenotype, bradycardia, angiogenesis inhibition and declined cardiac output in zebrafish co-exposed of SiNPs and PbAc at NOAEL level, significant changes were observed in mRNA and microRNA (miRNA) expression patterns. STC-GO analysis indicated that the co-exposure might have more toxic effects on cardiovascular system than that exposure alone. Key differentially expressed genes were discerned out based on the Dynamic-gene-network, including stxbp1a, ndfip2, celf4 and gsk3b. Furthermore, several miRNAs obtained from the miRNA-Gene-Network might play crucial roles in cardiovascular disease, such as dre-miR-93, dre-miR-34a, dre-miR-181c, dre-miR-7145, dre-miR-730, dre-miR-129-5p, dre-miR-19d, dre-miR-218b, dre-miR-221. Besides, the analysis of miRNA-pathway-network indicated that the zebrafish were stimulated by the co-exposure of SiNPs and PbAc, which might cause the disturbance of calcium homeostasis and endoplasmic reticulum stress. As a result, cardiac muscle contraction might be deteriorated. In general, our data provide abundant fundamental research clues to the combined toxicity of environmental pollutants and further in-depth verifications are needed.


Chemosphere | 2017

Gene expression profiles and bioinformatics analysis of human umbilical vein endothelial cells exposed to PM2.5

Hejing Hu; Collins Otieno Asweto; Jing Wu; Yanfeng Shi; Lin Feng; Xiaozhe Yang; Shuang Liang; Lige Cao; Junchao Duan; Zhiwei Sun

Cardiovascular system is demonstrated the main target of PM2.5 and the objective of this study was to explore the toxic effect and molecular mechanisms caused by PM2.5 in primary human umbilical vein endothelial cells (HUVECs) using microarray and bioinformatics analysis. The results showed that 591 genes were differentially expressed triggered by PM2.5, of which 174 genes were down-regulated, while 417 genes were up-regulated. Gene ontology analysis revealed that PM2.5 caused significant changes in gene expression patterns, including response to stimuli, immune response, and cellular processes. Pathway analysis and Signal-net analysis suggested that endocytosis, chemokine signaling pathway, RNA transport, protein processing in endoplasmic reticulum (ER) and autophagy regulation were the most critical pathways in PM2.5-induced toxicity in HUVECs. Moreover, gene expression confirmation of LIF, BCL2L1, CSF3, HMOX1, RPS6, PFKFB, CAPN1, HSPBP1, MOGS, PREB, TUBB2A, GABARAP by qRT-PCR indicated that endocytosis might be involved in the cellular uptake of PM2.5 by forming phagosomes, and subsequently inflammation, hypoxia and ER stress was occurred, which finally activated autophagy after PM2.5 exposure in HUVECs. In summary, our data can serve as fundamental research clues for further studies of PM2.5-induced toxicity in HUVECs.


Science of The Total Environment | 2019

PM2.5-induced alteration of DNA methylation and RNA-transcription are associated with inflammatory response and lung injury.

Yanfeng Shi; Tong Zhao; Xiaozhe Yang; Baiyang Sun; Yang Li; Junchao Duan; Zhiwei Sun

The mechanisms of systemic pulmonary inflammation and toxicity of fine particulate matter (PM2.5) exposure remains unclear. The current study investigated the inflammatory response and lung toxicity of PM2.5 in rats following intratracheal instillation of PM2.5. After repeated (treated every 3 days for 30 days) PM2.5 exposure, total protein (TP), lactate dehydrogenase (LDH) activity and inflammatory cytokines including interleukin 6 (IL-6), interleukin 1β (IL-1β) and tumor necrosis factor α (TNF-α) levels in bronchoalveolar lavage fluid (BALF) were markedly elevated. The expression levels of IL-6, IL-1β, TNF-α and NF-κB in rat lung tissue and BEAS-2B cells were significantly upregulated after PM2.5 exposure. Histopathological evaluation suggested that the major pathological changes were alveolar wall thickening and inflammatory cell infiltration of the lungs. Genome wide DNA methylation and RNA-transcription analysis was performed on human bronchial epithelial cells (BEAS-2B) to explore the potential mechanisms in vitro. PM2.5 induced genome wide DNA methylation and transcription changes. Differentially methylated CpGs were located in gene promoter region linked with CpG islands. Integrated analysis with DNA methylation and transcription data indicated a clear bias toward transcriptional alteration by differential methylation. Disease ontology of differentially methylated and expressed genes addressed their prominent role in respiratory disease. Functional enrichment revealed their involvement in inflammation or immune response, cellular community, cellular motility, cell growth, development and differentiation, signal transduction and responses to exogenous stimuli. Gene expression validation of ACTN4, CXCL1, MARK2, ABR, PSEN1, PSMA3, PSMD1 verified their functional participation in critical biological processes and supported the microarray bioinformatics analysis. Collectively, our data shows that PM2.5 induced genome wide methylome and transcriptome alterations that could be involved in pulmonary toxicity and pathological process of respiratory disease, providing new insight into the toxicity mechanisms of PM2.5.


Science of The Total Environment | 2018

Co-exposure of silica nanoparticles and methylmercury induced cardiac toxicity in vitro and in vivo

Xiaozhe Yang; Lin Feng; Yannan Zhang; Hejing Hu; Yanfeng Shi; Shuang Liang; Tong Zhao; Lige Cao; Junchao Duan; Zhiwei Sun

The released nanoparticles into environment can potentially interact with pre-existing pollution, maybe causing higher toxicity. As such, assessment of their joint toxic effects is necessary. This study was to investigate the co-exposure cardiac toxicity of silica nanoparticles (SiNPs) and methylmercury (MeHg). Factorial design was used to determine the potential joint action type. In vitro study, human cardiomyocytes (AC16) were exposed to SiNPs and MeHg alone or the combination. Higher toxicity was observed on cell viability, cell membrane damage in co-exposure compared with single exposure and control. The co-exposure enhanced the ROS, MDA generation and reduced the activity of SOD and GSH-Px. In addition, the co-exposure induced much higher cellular apoptotic rate in AC16. In vivo study, after SD rats exposed to SiNPs and MeHg and their mixture by intratracheal instillation for 30days, pathological changes (myocardial interstitial edema) of heart were occurred in co-exposure compared with single exposure and control. Moreover obvious ultra-structural changes, including myofibril disorder, myocardial gap expansion, and mitochondrial damage were observed in co-exposure group. The activity of myocardial enzymes, including CK-MB, ANP, BNP and cTnT, were significantly elevated in co-exposure group of rat serum. Meanwhile, the cardiac injury-linked proteins expression showed an increase in SERCA2 and decreased levels of cTnT, ANP and BNP in co-exposure group. Factorial design analysis demonstrated that additive and synergistic interactions were responsible for the co-exposure cardiac toxicity in vitro and vivo. In summary, our results showed severe cardiac toxicity induced by co-exposure of SiNPs and MeHg in both cardiomycytes and heart. It will help to clarify the potential cardiovascular toxicity in regards to combined exposure pollutions.


Ecotoxicology and Environmental Safety | 2018

DNA methylation: A critical epigenetic mechanism underlying the detrimental effects of airborne particulate matter

Baiyang Sun; Yanfeng Shi; Xiaozhe Yang; Tong Zhao; Junchao Duan; Zhiwei Sun

Exposure to airborne particulate matter (PM) does great harm to the health of human beings. To date, PM exposure has been closely associated with respiratory and cardiovascular diseases, as well as some types of cancer. As the associations of PM with the adverse health effects are well documented in literatures, the underlying mechanisms have not been completely clarified. With the field of epigenetics rising in recent years, PM-associated epigenetic alterations have gradually turned into the hot research topic. DNA methylation is one of the earliest-discovered and best-studied epigenetic mechanisms, of which the alteration can influence the transcription initiation of genes. A number of studies have been published to demonstrate that PM exposure is linked with DNA methylation patterns in the human genome. DNA methylation is the potential regulator of the biological effects of PM exposure. In the present review, DNA methylation related to PM exposure was elaborated on genome-wide and gene-specific methylation. In particular, genome-wide DNA methylation was composed of the alterations in global methylation content and genome-wide methylation profile; gene-specific methylation included the methylation changes in mechanism-related and disease-specific genes. Representative epidemiological and experimental studies were cited to elucidate the viewpoints, focusing on both PM-related methylation changes and the mediating effects of DNA methylation between PM and the health impacts. This review will provide advantageous clues for subsequent studies on the DNA methylation in relation to PM exposure.

Collaboration


Dive into the Yanfeng Shi's collaboration.

Top Co-Authors

Avatar

Junchao Duan

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhiwei Sun

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Lin Feng

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaozhe Yang

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Hejing Hu

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Yannan Zhang

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jing Wu

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Tong Zhao

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Shuang Liang

Capital Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge