Network


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

Hotspot


Dive into the research topics where Yu-He Yuan is active.

Publication


Featured researches published by Yu-He Yuan.


Neuropsychopharmacology | 2012

Gap Junction Dysfunction in the Prefrontal Cortex Induces Depressive-Like Behaviors in Rats

Jian-Dong Sun; Yan Liu; Yu-He Yuan; Jing Li; Nai-Hong Chen

Growing evidence has implicated glial anomalies in the pathophysiology of major depression disorder (MDD). Gap junctional communication is a main determinant of astrocytic function. However, it is unclear whether gap junction dysfunction is involved in MDD development. This study investigates changes in the function of astrocyte gap junction occurring in the rat prefrontal cortex (PFC) after chronic unpredictable stress (CUS), a rodent model of depression. Animals exposed to CUS and showing behavioral deficits in sucrose preference test (SPT) and novelty suppressed feeding test (NSFT) exhibited significant decreases in diffusion of gap junction channel-permeable dye and expression of connexin 43 (Cx43), a major component of astrocyte gap junction, and abnormal gap junctional ultrastructure in the PFC. Furthermore, we analyzed the effects of typical antidepressants fluoxetine and duloxetine and glucocorticoid receptor (GR) antagonist mifepristone on CUS-induced gap junctional dysfunction and depressive-like behaviors. The cellular and behavioral alterations induced by CUS were reversed and/or blocked by treatment with typical antidepressants or mifepristone, indicating that the mechanism of their antidepressant action may involve the amelioration of gap junction dysfunction and the cellular changes may be related to GR activation. We then investigated the effects of pharmacological gap junction blockade in the PFC on depressive-like behaviors. The results demonstrate that carbenoxolone (CBX) infusions induced anhedonia in SPT, and anxiety in NSFT, and Cx43 mimetic peptides Gap27 and Gap26 also induced anhedonia, a core symptom of depression. Together, this study supports the hypothesis that gap junction dysfunction contributes to the pathophysiology of depression.


European Journal of Pharmacology | 2013

Ginsenoside Rg1 attenuates okadaic acid induced spatial memory impairment by the GSK3β/tau signaling pathway and the Aβ formation prevention in rats.

Xiu-Yun Song; Jin-Feng Hu; Shi-feng Chu; Zhao Zhang; Shuang Xu; Yu-He Yuan; Ning Han; Yan Liu; Fei Niu; Xin He; Nai-Hong Chen

Ginsenoside Rg1, one of the major active ingredients isolated from Panax Ginseng, has been shown notable neuroprotective effects in memory impairment animals. However, the role of ginsenoside Rg1 on cognition capacity damaged by neurofibrillary tangles (NFTs) is still poorly understood, and the underlying mechanism remain to be fully elucidated. Okadaic acid (OKA), a potent phosphatase inhibitor, often apply to imitate Alzheimers disease-like symptom damaged by neurofibrillary tangles, was used to investigate the effects of ginsenoside Rg1 on memory impairment and the related mechanisms in Sprague Dawley (SD) rats. The anti-dementic drug donepezil was used as a positive contrast. The results showed that OKA intracerebroventricular (i.c.v.) injection induced memory impairment, including changes in the ability of orientation navigate, spatial probe and relearning memory in behavioral test of Morris water maze (MWM). However, treatment with Rg1 and donepezil remarkably alleviated these changes. Also OKA treated rats showed memory impairment including increasing of phospho-tau, decreasing of phospho-GSK3β and the formation of β-amyloid in special brain regions, which were reversed by Rg1 (20 mg/kg) and donepezil (1 mg/kg) administration. All these indicating that ginsenoside Rg1 protects rats against OKA-induced neurotoxicity. The possible neuroprotective mechanism may be that Rg1 decreases OKA-induced memory impairment through GSK3β/tau signaling pathway and/or attenuating Aβ formation. Thus, these studies indicate that ginsenoside Rg1 might be a potential preventive drug for Alzheimers disease.


Journal of Natural Products | 2011

Bioactive neolignans and lignans from the bark of Machilus robusta.

Yanru Li; Wei Cheng; Chenggen Zhu; Chun-Suo Yao; Liang Xiong; Ye Tian; S. Wang; Sheng Lin; Jinfeng Hu; Yongchun Yang; Ying Guo; Ying Yang; Yan Li; Yu-He Yuan; Nai-Hong Chen; Jiangong Shi

Sixteen new neolignans and lignans (1-16), together with 12 known analogues, have been isolated from an ethanol extract of the bark of Machilus robusta. Compounds 1 and 2 showed activity against HIV-1 replication in vitro, with IC(50) values of 2.52 and 2.01 μM, respectively. At 10 μM, 6, 8, and 9 reduced dl-galactosamine-induced hepatocyte (WB-F344 cells) damage, and 9 could additionally attenuate rotenone-induced PC12 cell damage. The known compounds (-)-pinoresinol (17) and (+)-lyoniresinol (18) were active against serum deprivation induced PC12 cell damage.


Cellular and Molecular Neurobiology | 2008

Overexpressed alpha-synuclein regulated the nuclear factor-kappaB signal pathway.

Yu-He Yuan; Jin Jin; Bo Yang; Wei Zhang; Jin-feng Hu; Yun Zhang; Nai-Hong Chen

Alpha-synuclein is a presynaptic protein which is implicated in some neurodegenerative disorders including Parkinson’s disease, dementia with Lewy bodies, multiple systems atrophy, and Hallervorden-Spatz disease, and its overexpression contributes to the loss of dopaminergic neurons. Although the role of alpha-synuclein in these paradigms has been widely documented, its exact function is still elusive. And the dysfunction of the transcription factor nuclear factor (NF-κB) also exists in many neurodegenerative diseases. In this reason the purpose of this study was to investigate the molecular mechanism of alpha-synuclein’s toxicity and its association with NF-κB by MTT assay, Western blot method, and luciferase assay. Results showed that overexpressed alpha-synuclein and 1-methyl-4-phenylpyridinium (MPP+) suppressed the SH-SY5Y cell viability and attenuate NF-κB-mediated luciferase expression significantly. Moreover, the impairment function was enhanced with the increase of alpha-synuclein protein level. We also found that overexpressed alpha-synuclein localized both in the cytoplasms and nuclei, down-regulated the anti-apoptotic Bcl-2 expression and up-regulated the pro-apoptotic glycogen synthase kinase 3β (GSK3β) protein level. In conclusion, all these findings mentioned above suggested that alpha-synuclein shared some toxic functional homology with neurotoxin MPP+, and the proapoptotic effects of alpha-synuclein might be mediated at least in part by the impairment of NF-κB signaling pathway which involves GSK3β.


Journal of Medicinal Chemistry | 2010

Discovery and Optimization of Novel 3-Piperazinylcoumarin Antagonist of Chemokine-like Factor 1 with Oral Antiasthma Activity in Mice

Gang Li; Dongmei Wang; Mingna Sun; Guangyan Li; Jin-Feng Hu; Yun Zhang; Yu-He Yuan; Haijie Ji; Nai-Hong Chen; Gang Liu

Chemokine-like factor 1 (CKLF1) is a novel functional cytokine that acts through its receptor CC chemokine receptor 4 (CCR4). Activation of CCR4 by CKLF1 plays an important role in diseases such as asthma and multiple sclerosis. This article describes a cell-based screening assay using an FITC-labeled CCR4 agonist (CKLF1-C27), a CKLF1 peptide fragment. Screening of our in-stock small-molecule library identified a 3-piperazinylcoumarin analogue 1 (IC(50) = 4.36 x 10(-6) M) that led to the discovery of orally active compound 41 (IC(50) = 2.12 x 10(-8) M) through systematic optimization. Compound 41 blocked the calcium mobilization and chemotaxis induced by CKLF1-C27 and reduced the asthmatic pathologic changes in lung tissue of human CKLF1-transfected mice. Further studies indicated that compound 41 ameliorated pathological changes via inhibition of the NF-kappaB signal pathway.


Molecular Neurobiology | 2014

Overexpression of Human E46K Mutant α-Synuclein Impairs Macroautophagy via Inactivation of JNK1-Bcl-2 Pathway

Jia-qing Yan; Yu-He Yuan; Yan-Na Gao; Ju-yang Huang; Kai-Li Ma; Yan Gao; Wanqing Zhang; Xiao-Feng Guo; Nai-Hong Chen

Parkinson’s disease (PD) is pathologically characterized by selective loss of dopaminergic neurons in the midbrain and the existence of intracellular protein inclusions termed Lewy bodies, largely composed of α-synuclein. Genetic studies have revealed that rare point mutations in the gene encoding α-synuclein including A30P, A53T, and E46K are associated with familial forms of PD, indicating a pathological role for mutant α-synuclein in PD etiology. However, the mechanisms underlying the neuronal toxicity of mutant α-synuclein are still to be elucidated. Growing evidence has suggested a deleterious effect of mutant α-synuclein on the autophagy-lysosome pathway. In this study, we discovered that overexpression of human E46K mutant α-synuclein impaired macroautophagy in mammalian cells. Our data showed that overexpression of E46K mutant α-synuclein impaired autophagy at an early stage of autophagosome formation via the c-Jun N-terminal kinase 1 (JNK1)-Bcl-2 but not the mammalian target of rapamycin (mTOR) pathway. Overexpressed E46K mutant α-synuclein inhibited JNK1 activation, leading to a reduced Bcl-2 phosphorylation and increased association between Bcl-2 and Beclin1, further disrupting the formation of Beclin1/hVps34 complex, which is essential for autophagy initiation. Furthermore, overexpression of E46K mutant α-synuclein increased the vulnerability of differentiated PC12 cells to rotenone treatment, which would be partly due to its inhibitory effects on autophagy. Our findings may shed light on the potential roles of mutant α-synuclein in the pathogenesis of PD.


Brain Research | 2011

Inhibitory effect of ginsenoside Rg1 on lipopolysaccharide-induced microglial activation in mice.

Jin-Feng Hu; Xiu-Yun Song; Shi-feng Chu; Ji Chen; Haijie Ji; Xiao-Yu Chen; Yu-He Yuan; Ning Han; Jun-Tian Zhang; Nai-Hong Chen

Microglial activation plays a pivotal role in the pathogenesis of neurodegenerative diseases by producing various pro-inflammatory cytokines and nitric oxide (NO). In the paper, the anti-inflammatory effect of ginsenoside Rg1 was investigated in mice intracerebroventricular injected of lipopolysaccharide (LPS). NO and tumor necrosis factor (TNF)-α production in both cerebral cortex and hippocampus decreased at dose-dependent manner by oral administration with Rg1. And the expression of ionized calcium binding adaptor molecule 1 (Iba-1) increased in both cerebral cortex and hippocampus in LPS-injected group compared to that in control group. However, Rg1 inhibited microglial activation by suppressing Iba-1 expression. In addition, the expression of inducible nitric oxide synthase (iNOS) was inhibited by Rg1. Moreover, Rg1 suppressed the phosphorylation level of IκB, nuclear translocation of p65 subunit of NFκB, and phosphorylation level of p38, ERK1/2, JNK mitogen-activated protein kinase (MAPK) induced by LPS. Concluding, Rg1 inhibited the inflammation mediated by LPS by suppressing NFκB and MAPK pathway, which provided the explanation for its therapeutic effect on neurodegenerative diseases.


Toxicology Letters | 2016

Ginsenoside Rg1 attenuates motor impairment and neuroinflammation in the MPTP-probenecid-induced parkinsonism mouse model by targeting α-synuclein abnormalities in the substantia nigra.

Yang Heng; Qiu-Shuang Zhang; Zheng Mu; Jin-Feng Hu; Yu-He Yuan; Nai-Hong Chen

Parkinsons disease (PD) is pathologically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the accumulation of aggregated α-synuclein in specific central nervous system (CNS) regions. Disease development is attributed to α-synuclein abnormalities, particularly aggregation and phosphorylation. The ginsenoside Rg1, an active component of ginseng, possesses neuroprotective and anti-inflammatory effects. The purpose of the present study was to evaluate these activities of Rg1 in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid (MPTP/p)-induced PD mouse model for the first time and to elucidate the underlying mechanisms. Oral treatment with Rg1 significantly attenuated the high MPTP-induced mortality, behavior defects, loss of dopamine neurons and abnormal ultrastructure changes in the SNpc. Other assays indicated that the protective effect of Rg1 may be mediated by its anti-neuroinflammatory properties. Rg1 regulated MPTP-induced reactive astrocytes and microglia and decreased the release of cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the SNpc. Rg1 also alleviated the unusual MPTP-induced increase in oligomeric, phosphorylated and disease-related α-synuclein in the SNpc. In conclusion, Rg1 protects dopaminergic neurons, most likely by reducing aberrant α-synuclein-mediated neuroinflammation, and holds promise for PD therapeutics.


Toxicology Letters | 2015

The molecular mechanism of rotenone-induced α-synuclein aggregation: Emphasizing the role of the calcium/GSK3β pathway

Yu-He Yuan; Wen-Fen Yan; Jian-Dong Sun; Ju-yang Huang; Zheng Mu; Nai-Hong Chen

Environmental toxin exposure is associated with the development of Parkinsons disease (PD), and environmental factors can influence the onset of the majority of sporadic PD cases via genetically mediated pathways. Rotenone, a widespread pesticide, induces Parkinsonism and the formation of Lewy bodies in animals; however, the molecular mechanism that underlies α-synuclein aggregation remains unclear. Here, we assessed the aggregation of α-synuclein in PC12 cells with or without cross-linking following rotenone exposure via a variety of methods, including western blotting, immunofluorescence and electron microscopy. We demonstrated that rotenone increased the intracellular calcium levels and induced the aggregation and phosphorylation of α-synuclein in a calcium-dependent manner. Aggregated α-synuclein is typically degraded by autophagy, and rotenone impaired this process. The attenuation of autophagy and α-synuclein alterations were reversed by scavenging calcium. Calcium regulates the activity of AKT-glycogen synthase kinase 3 (GSK3)β. We demonstrated that rotenone attenuated the phosphorylation of AKT and GSK3β, and the elimination of calcium reversed these phenomena. As a GSK3β inhibitor, lithium promoted autophagy and decreased the aggregation and phosphorylation of α-synuclein. GSK3β activation through overexpression depressed autophagy and increased the total protein level and phosphorylation of α-synuclein. These results suggest that rotenone-induced α-synuclein aggregation is mediated by the calcium/GSK3β signaling pathway.


Neuroscience Letters | 2010

Protective effect of (−)clausenamide against Aβ-induced neurotoxicity in differentiated PC12 cells

Jin-feng Hu; Shi-feng Chu; Na Ning; Yu-He Yuan; Wei Xue; Nai-Hong Chen; Jun-Tian Zhang

The neurotoxicity of aggregated beta-amyloid (Abeta) has been implicated as a critical cause in the pathogenesis of Alzheimers disease (AD). In the present study, we investigated the effect of (-)clausenamide ((-)Clau), an aqueous extract of leaves of Clausena lassium (lour) skeel, on the neurotoxicity of Abeta(25-35). The viability of differentiated PC12 cells was determined by MTT assay. Apoptosis was detected by flow cytometry. DCFH-DA was used for assessment of intracellular ROS generation, JC-1 and Rhodamine 123 for measurement of mitochondrial transmembrane potential (MMP). The intracellular calcium was determined with Fluo-3. The phosphorylation of p38 MAPK and the expression of Bcl-2, Bax, P53, Caspase 3 were examined by Western blot. The results showed that (-)Clau significantly elevated cell viability. Furthermore, (-)Clau arrested the apoptotic cascade by reversing overload of calcium, preventing ROS generation, moderated the dissipation of MMP and the misbalance of Bcl-2 and Bax, inhibiting the activation of p38 MAPK and the expression of P53 and cleaved Caspase 3. Our results suggested that (-)Clau may be a therapeutic agent for AD.

Collaboration


Dive into the Yu-He Yuan's collaboration.

Top Co-Authors

Avatar

Nai-Hong Chen

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jin-Feng Hu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Ning Han

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Kai-Li Ma

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Qian-hang Shao

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Shi-feng Chu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Dong-Ming Zhang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Lian-Kun Song

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Chenggen Zhu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jian-Dong Sun

Peking Union Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge