Network


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

Hotspot


Dive into the research topics where Xiaoli Yao is active.

Publication


Featured researches published by Xiaoli Yao.


Journal of Lipid Research | 2014

Enriched endogenous omega-3 fatty acids in mice protect against global ischemia injury

Chuanming Luo; Huixia Ren; Jian-Bo Wan; Xiaoli Yao; Xiao-Jing Zhang; Chengwei He; Kf So; Jing X. Kang; Zhong Pei; Huanxing Su

Transient global cerebral ischemia, one of the consequences of cardiac arrest and cardiovascular surgery, usually leads to delayed death of hippocampal cornu Ammonis1 (CA1) neurons and cognitive deficits. Currently, there are no effective preventions or treatments for this condition. Omega-3 (ω-3) PUFAs have been shown to have therapeutic potential in a variety of neurological disorders. Here, we report that the transgenic mice that express the fat-1 gene encoding for ω-3 fatty acid desaturase, which leads to an increase in endogenous ω-3 PUFAs and a concomitant decrease in ω-6 PUFAs, were protected from global cerebral ischemia injury. The results of the study show that the hippocampal CA1 neuronal loss and cognitive deficits induced by global ischemia insult were significantly less severe in fat-1 mice than in WT mice controls. The protection against global cerebral ischemia injury was closely correlated with increased production of resolvin D1, suppressed nuclear factor-kappa B activation, and reduced generation of pro-inflammatory mediators in the hippocampus of fat-1 mice compared with WT mice controls. Our study demonstrates that fat-1 mice with high endogenous ω-3 PUFAs exhibit protective effects on hippocampal CA1 neurons and cognitive functions in a global ischemia injury model.


Brain Research | 2010

Protective effects of xyloketal B against MPP+-induced neurotoxicity in Caenorhabditis elegans and PC12 cells.

Xilin Lu; Xiaoli Yao; Zhi-yong Liu; Heng Zhang; Wei Li; Zhenxing Li; Guan-Lei Wang; Jiyan Pang; Yongcheng Lin; Zhongliang Xu; Ling Chen; Zhong Pei; Jinsheng Zeng

Parkinsons disease (PD) is the second most common neurodegenerative disease, affecting 2% of the population over age 65years. Mitochondrial defect and oxidative stress actively participate in the dopaminergic (DA) neuron degeneration in PD. Xyloketal B is a novel marine compound with unique chemical structure isolated from mangrove fungus Xylaria sp. (no. 2508). Recently, we have demonstrated that Xyloketal B can directly scavenge DPPH free radicals and protects mitochondria against oxidative insult. In the present study, we investigate the neuroprotective action of xyloketal B against MPP+-induced neurotoxicity in Caenorhabditis elegans and PC12 cells. The viability and DA neurodegeneration was assessed in C. elegans selectively expressing green fluorescent protein (GFP) in DA neurons. PC12 cell damage was measured using MTT and nuclear morphology. Intracellular reactive oxygen species (ROS), mitochondrial membrane potential and total GSH were assessed. Xyloketal B dose-dependently protected against MPP+-induced loss of viability and DA neurodegeneration in C. elegans. Similar neuroprotection was replicated in MPP+ PC12 cell model. In addition, xyloketal B attenuated MPP+-induced intracellular ROS accumulation, loss of mitochondrial membrane potential and restored total GSH level in PC12 cells. All together, the present study demonstrates that xyloketal B protects against MPP+-induced neurotoxicity in C. elegans and PC12 cells mainly through its antioxidant property and restoration of total GSH level.


Stem Cell Research | 2013

Immediate expression of Cdh2 is essential for efficient neural differentiation of mouse induced pluripotent stem cells.

Huanxing Su; Lihui Wang; Wenhao Huang; Dajiang Qin; Jinglei Cai; Xiaoli Yao; Chengqian Feng; Zhiyuan Li; Wang Y; Kf So; Guangjin Pan; Wutian Wu; Duanqing Pei

Induced pluripotent stem cells (iPSCs) exhibit reduced efficiency and higher variability in neural differentiation compared to embryonic stem cells (ESCs). In this study, we showed that mouse iPSCs failed to efficiently give rise to neuronal cells using conventional methods previously established for driving mouse ESC differentiation. We reported a novel approach which remarkably increases neural differentiation of mouse iPSCs. This novel approach initiated embryoid body (EB) formation directly from the whole cell clones isolated from the top of feeder cells. Compared to conventional neural induction methods such as single cell suspension or monolayer culture, the cell clone-derived EB method led to a pronounced increase in directed generation of various types of neural cells including neural stem cells, motoneurons and dopaminergic neurons in response to different inducers. Through gene expression microarray analysis, we identified 14 genes that were highly expressed in the cell clone-derived EBs. Among them, we found that Cdh2, also known as N-cadherin, played important roles in controlling the neural differentiation efficiency of mouse iPSCs. Forced expression of Cdh2 in iPSCs substantially enhanced the differentiation efficiency while knocking-down of Cdh2 by shRNA blocked the neural differentiation. Our results revealed a critical role of Cdh2 in the process of efficient neural differentiation of mouse iPS cells.


PLOS ONE | 2013

Motoneuron Differentiation of Induced Pluripotent Stem Cells from SOD1G93A Mice

Xiaoli Yao; Cheng-Hui Ye; Qiang Liu; Jian-Bo Wan; Jun Zhen; Andy Peng Xiang; Weiqiang Li; Wang Y; Huangxing Su; Xilin Lu

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder mainly affecting motor neurons. Mutations in superoxide dismutase-1 (SOD-1) account for about 20% of familial ALS patients. A robust supply of motoneurons carrying the mutated gene would help understand the causes of motoneuron death and develop new therapeutics for the disease. Here, we established induced pluripotent stem (iPS) cell lines from SOD1G93A mice and compared their potency in motoneuron generation with normal iPS cells and mouse embryonic stem cells (E14). Our results showed that iPS cells derived from SOD1G93A mice possessed the similar potency in neuronal differentiation to normal iPS cells and E14 cells and can be efficiently driven to motoneuron-like phenotype. These cells exhibited typical neuronal morphology, expressed key motoneuron markers, including ChAT and HB9, and generated repetitive trains of action potentials. Furthermore, these neurons highly expressed human SOD-1 and exhibited shorter neurites compared to controls. The present study provides evidence that ALS-iPS cells can be used as disease models in high-throughput screening and mechanistic studies due to their ability to efficiently differentiate into specific neuronal subtypes.


Journal of Cardiovascular Pharmacology | 2012

dl-3n-Butylphthalide promotes angiogenesis via the extracellular signal-regulated kinase 1/2 and phosphatidylinositol 3-kinase/Akt-endothelial nitric oxide synthase signaling pathways.

Xilin Lu; Dan Luo; Xiaoli Yao; Guang-lei Wang; Zhi-yong Liu; Zhenxing Li; Wei Li; Feng-Jun Chang; Lu Wen; Simon Ming-Yuen Lee; Zaijun Zhang; Ling Li; Jinsheng Zeng; Ruxun Huang; Zhong Pei; Jing-Song Ou

Abstract: We have previously demonstrated that dl-3n-butylphthalide (NBP) has a potential angiogenic activity. In this study, we investigated the angiogenic effect of NBP and the molecular mechanisms underlying NBP-mediated angiogenesis. Zebrafish embryos and human umbilical vein endothelial cells were treated with various doses of NBP and several signaling pathway inhibitors. NBP induced ectopic subintestinal vessel production in zebrafish embryos and induced invasion, migration, and endothelial cell tube formation of human umbilical vein endothelial cells in a dose-dependent manner. These NBP-induced angiogenic effects were partially suppressed by SU5402, a fibroblast growth factor receptor 1 inhibitor; U0126, an extracellular signal–regulated kinase 1/2 (ERK1/2) inhibitor; LY294002, a phosphatidylinositol 3-kinase inhibitor; 1L6-hydroxymethyl-chiro-inositol-2-(R)-2-O-methyl-3-O-octadecyl-sn-glycerocarbonate, an Akt inhibitor; cavtratin, an endothelial nitric oxide synthase (eNOS) inhibitor and completely inhibited by a combination of U0126 and LY294002. NBP enhanced phosphorylation of ERK1/2 and fibroblast growth factor receptor 2 expression, which were inhibited by U0126. NBP increased the phosphorylation of Akt and eNOS at serine 1177, which was blocked by LY294002. NBP-stimulated nitric oxide production, which was reduced by LY294002. Our data demonstrated that (1) NBP promoted angiogenesis and (2) the angiogenic effects of NBP were mediated by the ERK1/2 and phosphatidylinositol 3-kinase/Akt-eNOS signaling pathways. Our findings suggest that NBP could be a novel agent for therapeutic angiogenesis in ischemic diseases.


Molecular Neurobiology | 2016

Enriched Endogenous Omega-3 Polyunsaturated Fatty Acids Protect Cortical Neurons from Experimental Ischemic Injury

Zhe Shi; Huixia Ren; Chuanming Luo; Xiaoli Yao; Peng Li; Chengwei He; Jing-X Kang; Jian-Bo Wan; Ti-Fei Yuan; Huanxing Su

Omega-3 polyunsaturated fatty acids (n-3 PUFAs) exert therapeutic potential in a variety of neurological disorders, including ischemic stroke. However, the underlying mechanisms still lack investigation. Here, we report that cultured cortical neurons isolated from fat-1 mice with high endogenous n-3 PUFAs were tolerant to oxygen-glucose deprivation/reperfusion (OGD/R) injury. Fat-1 neurons exhibited significantly attenuated reactive oxygen species (ROS) activation induced by OGD/R injury, upregulated antiapoptotic proteins Bcl-2 and Bcl-xL, and reduced cleaved caspase-3. Exogenous administration of docosahexaenoic acid (DHA), a major component of the n-3 PUFA family, resulted in similar protective effects on cultured cortex neurons. We further verified the protective effects of n-3 PUFAs in vivo, using a mini ischemic model with a reproducible cortical infarct and manifest function deficits by occlusion of the distal branch of the middle cerebral artery with focused femtosecond laser pulses. The Fat-1 animals showed decreased ROS expression and higher level of glutathione in the injured brain, associated with improved functional recovery. We therefore provide evidence that n-3 PUFAs exert their protective effects against ischemic injury both in vitro and in vivo, partly through inhibiting ROS activation.


The FASEB Journal | 2017

Omega-3 polyunsaturated fatty acids promote amyloid-β clearance from the brain through mediating the function of the glymphatic system

Huixia Ren; Chuanming Luo; Yanqing Feng; Xiaoli Yao; Zhe Shi; Fengyin Liang; Jing X. Kang; Jian-Bo Wan; Zhong Pei; Huanxing Su

Impairment of amyloid‐β (Aβ) clearance leads to Aβ accumulation in the brain during the development of Alzheimers disease (AD). Strategies that can restore or improve the clearance function hold great promise in delaying or preventing the onset of AD. Here, we show that n‐3 polyunsaturated fatty acids (PUFAs), by use of fat‐1 transgenicmice andoral administration of fish oil, significantly promote interstitial Aβ clearance from theb rain and resist Aβ injury. Such beneficial effects were abolished in Aqp4‐knockout mice, suggesting that the AQP4‐ dependent glymphatic system is actively involved in the promoting the effects of n‐3 PUFAs on the clearance of extracellularAβ. Imaging on clarified brain tissues clearly displayed thatn‐3 PUFAsmarkedly inhibit the activation of astrocytes and protect the AQP4 polarization in the affected brain region after Aβ injection. The results of the present study prove a novelmechanism by which n‐3 PUFAs exert protective roles in reducingAβ accumulation via mediating the glymphatic system function.—Ren, H., Luo, C., Feng, Y., Yao, X., Shi, Z., Liang, F., Kang, J. X., Wan, J.‐B., Pei, Z., Su, H. Omega‐3 polyunsaturated fatty acids promote amyloid‐β clearance from the brain through mediating the function of the glymphatic system. FASEB J. 31, 282–293 (2017) www.fasebj.org


Molecular Medicine Reports | 2016

Neuroprotective effects of ginsenosides on neural progenitor cells against oxidative injury

Jun Ye; Jian‑Ping Yao; Xu Wang; Minying Zheng; Peng Li; Chengwei He; Jian-Bo Wan; Xiaoli Yao; Huanxing Su

Ginsenosides exhibit various neuroprotective effects against oxidative stress. However, which ginsenoside provides optimal effects for the treatment of neurological disorders as a potent antioxidant remains to be elucidated. Therefore, the present study investigated and compared the neuroprotective effects of the Rb1, Rd, Rg1 and Re ginsenosides on neural progenitor cells (NPCs) following tert-Butylhydroperoxide (t-BHP)-induced oxidative injury. Primary rat embryonic cortical NPCs were prepared from E14.5 embryos of Sprague-Dawley rats. The oxidative injury model was established with t-BHP. A lactate dehydrogenase assay and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining were used to measure the viability of the NPCs pre-treated with ginsenosides under oxidative stress. Reverse transcription-quantitative polymerase chain reaction analysis was used to determine the activation of intracellular signaling pathways triggered by the pretreatment of ginsenosides. Among the four ginsenosides, only Rb1 attenuated t-BHP toxicity in the NPCs, and the nuclear factor (erythroizd-derived 2)-like 2/heme oxygenase-1 pathway was found to be key in the intracellular defense against oxidative stress. The present study demonstrated the anti-oxidative effects of ginsenoside Rb1 on NPCs, and suggested that Rb1 may offer potential as a potent antioxidant for the treatment of neurological disorders.


Molecular Neurobiology | 2016

Endogenous Docosahexaenoic Acid (DHA) Prevents Aβ1–42 Oligomer-Induced Neuronal Injury

Yuan Tan; Huixia Ren; Zhe Shi; Xiaoli Yao; Chengwei He; Jing-X Kang; Jian-Bo Wan; Peng Li; Ti-Fei Yuan; Huanxing Su

The intake of the polyunsaturated fatty acid docosahexaenoic acid (DHA) or n-3 fatty acid has been associated with reduced risk of Alzheimer’s disease (AD) in epidemiological reports. However, the underlying mechanism remains to be elucidated. Here, we report that exogenous DHA administration could protect neurons against Aβ oligomer-induced injury both in vitro and in vivo, partly through reducing the endoplasmic reticulum (ER) stress, and preventing cell apoptosis. In transgenic fat-1 mice with enriched ω-3 fatty acids, Aβ oligomers induced fewer neuronal losses, when compared to wild-type (WT) mice. We conclude that endogenous DHA are neuroprotective in pathogenesis processes of AD.


Marine Drugs | 2014

Marine Compound Catunaregin Inhibits Angiogenesis through the Modulation of Phosphorylation of Akt and eNOS in vivo and in vitro

Jun-Xiu Liu; Min-Qi Luo; Meng Xia; Qi Wu; Simei Long; Yaohua Hu; Guang-Chun Gao; Xiaoli Yao; Mian He; Huanxing Su; Xiong-Ming Luo; Shuzhong Yao

Angiogenesis is the formation of blood vessels from pre-existing vasculature. Excessive or uncontrolled angiogenesis is a major contributor to many pathological conditions whereas inhibition of aberrant angiogenesis is beneficial to patients with pathological angiogenesis. Catunaregin is a core of novel marine compound isolated from mangrove associate. The potential anti-angiogenesis of catunaregin was investigated in human umbilical vein endothelial cells (HUVECs) and zebrafish. HUVECs were treated with different concentrations of catunaregin in the presence or absence of VEGF. The angiogenic phenotypes including cell invasion cell migration and tube formation were evaluated following catunaregin treatment in HUVECs. The possible involvement of AKT, eNOS and ERK1/2 in catunaregin-induced anti-angiogenesis was explored using Western blotting. The anti-angiogenesis of catunaregin was further tested in the zebrafish embryo neovascularization and caudal fin regeneration assays. We found that catunaregin dose-dependently inhibited angiogenesis in both HUVECs and zebrafish embryo neovascularization and zebrafish caudal fin regeneration assays. In addition, catunaregin significantly decreased the phosphorylation of Akt and eNOS, but not the phosphorylation of ERK1/2. The present work demonstrates that catunaregin exerts the anti-angiogenic activity at least in part through the regulation of the Akt and eNOS signaling pathways.

Collaboration


Dive into the Xiaoli Yao's collaboration.

Top Co-Authors

Avatar

Zhong Pei

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Xilin Lu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Baixuan He

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ti-Fei Yuan

Nanjing Normal University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Simei Long

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qi Wu

Sun Yat-sen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge