Network


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

Hotspot


Dive into the research topics where Nan Feng is active.

Publication


Featured researches published by Nan Feng.


Journal of Alzheimer's Disease | 2012

L-3-n-butylphthalide reduces tau phosphorylation and improves cognitive deficits in AβPP/PS1-Alzheimer's transgenic mice.

Ying Peng; Yanli Hu; Shaofeng Xu; Pingping Li; Jiang Li; Li Lu; Hongyan Yang; Nan Feng; Ling Wang; Xiaoliang Wang

L-3-n-butylphthalide (L-NBP), an extract from seeds of Apium graveolens Linn (Chinese celery), has been shown to have neuroprotective effects on cerebral ischemic, vascular dementia and amyloid-β (Aβ)-induced animal models by inhibiting oxidative injury, neuronal apoptosis and glial activation, regulating amyloid-β protein precursor (AβPP) processing and reducing Aβ generation. The aim of the present study was to examine the effect of L-NBP on learning and memory in AβPP and presenilin 1 (PS1) double-transgenic AD mouse model (AβPP/PS1) and the mechanisms of L-NBP in reducing Aβ accumulation and tau phosphorylation. Twelve-month old AβPP/PS1 mice were given 15 mg/kg L-NBP by oral gavage for 3 months. L-NBP treatment significantly improved the spatial learning and memory deficits compared to the vehicle-treated AβPP/PS1 mice, whereas L-NBP treatment had no effect on cerebral Aβ plaque deposition and Aβ levels in brain homogenates. However, we found an L-NBP-induced reduction of tau hyperphosphorylation at Ser199, Thr205, Ser396, and Ser404 sites in AβPP/PS1 mice. Additionally, the expressions of cyclin-dependent kinase and glycogen synthase kinase 3β, the most important kinases involved in tau phosphorylation, were markedly decreased by L-NBP treatment. The effects of L-NBP on decreasing tau phosphorylation and kinases activations were further confirmed in neuroblastoma SK-N-SH cells overexpressing wild-type human AβPP695 (SK-N-SH AβPPwt). L-NBP shows promising candidate of multi-target neuronal protective agent for the treatment of Alzheimers disease.


European Journal of Pharmacology | 2011

Donepezil attenuated oxygen-glucose deprivation insult by blocking Kv2.1 potassium channels.

Hui Yuan; Weiping Wang; Nan Feng; Ling Wang; Xiaoliang Wang

Excessive K(+) efflux via activated voltage-gated K(+) channels (Kv channels) and the consequent intracellular K(+) depletion during and after ischemia/hypoxia induced long-lasting membrane depolarization promotes neuronal apoptosis. Although it has been suggested as an important potassium channel subtype in oxidative stress induced neuron apoptosis, whether Kv2.1 mediates ischemic apoptosis remains undefined. In the present study, the role of Kv2.1 played in hypoxia/anoxia induced cell apoptosis and correlated protective effect of donepezil were evaluated. Kv2.1 transfected HEK293 cell line (Kv2.1/HEK293) was used to study oxygen-glucose deprivation (OGD) induced cell apoptosis. We found Kv2.1 transfection increased the vulnerability of HEK293 cells to OGD insult, blocking Kv2.1 potassium channel by tetraethylammonium (TEA, 10mM) could attenuated OGD induced Kv2.1/HEK293 cell apoptosis significantly. OGD slightly reduced Kv2.1 currents without affecting channel kinetic activity. However, the membrane potential of Kv2.1/HEK293 cells depolarized to around 0mV after OGD treatment, a potential which could activated Kv2.1 persistently. Donepezil blocked Kv2.1 currents in a dose-dependent manner (IC(50)=7.59μM). Under OGD condition, donepezil (30μM) effectively inhibited Kv2.1 currents by accelerating channel inactivation and decreased Kv2.1/HEK293 cell apoptosis rate. In conclusion, our study revealed both the conducting role of Kv2.1 in OGD induced cell apoptosis and the importance of Kv2.1 as a target for neuronal protection. In addition, besides anti-acetylcholinesterase activity, Kv2.1 blockade capability of donepezil may attribute to its neuroprotective effects against ischemic apoptosis.


Neuroscience Letters | 2011

L-3-n-butylphthalide regulates amyloid precursor protein processing by PKC and MAPK pathways in SK-N-SH cells over-expressing wild type human APP695.

Ying Peng; Yanli Hu; Shaofeng Xu; Nan Feng; Ling Wang; Xiaoliang Wang

Amyloid precursor protein (APP) is cleaved by α-secretase, within the amyloid-β (Aβ) sequence, resulting in the release of a secreted fragment (αAPPs) and precluding Aβ production. We investigated the effects of a promising anti-AD new drug, l-3-n-butylphthalide (L-NBP), on APP processing and Aβ generation in neuroblastoma SK-N-SH cells overexpressing wild-type human APP695. L-NBP significantly increased αAPPs release, and reduced Aβ generation. The steady-state full-length APP levels were unaffected by L-NBP. It suggested that L-NBP regulated APP processing towards to the non-amyloidogenic α-secretase pathway. Protein kinase C (PKC) and mitogen activated protein (MAP) kinase might be involved in L-NBP-induced αAPPs secretion. L-NBP significantly increased PKCα and ɛ activations, lowered PKCγ activation and increased the phosphorylation of p44/p42 MAPK. Furthermore, PKC and MAPK inhibitors partially reduced L-NBP-induced αAPPs secretion. The results suggested alternative pharmacological mechanisms of L-NBP regarding the treatment of Alzheimers disease (AD).


Journal of Cardiovascular Pharmacology | 2013

An increased TREK-1-like potassium current in ventricular myocytes during rat cardiac hypertrophy.

Weiping Wang; Man Zhang; Pingping Li; Hui Yuan; Nan Feng; Ying Peng; Ling Wang; Xiaoliang Wang

Abstract: To elucidate the expression and identify the functional changes of 2 pore domain potassium channel TREK-1 during cardiac hypertrophy in rats, left ventricular hypertrophy was induced by subcutaneous injection with isoproterenol. Western blot was used to detect the expression of TREK-1 channel protein, and inside-out and whole-cell recordings were used to record TREK-1 currents. The results showed that TREK-1 protein expression in endocardium was slightly higher than that in epicardium in control left ventricles. However, it was obviously upregulated by 89.8% during hypertrophy, 2.3-fold higher than in epicardium. Mechanical stretch, intracellular acidification, and arachidonic acid could activate a TREK-1–like current in cardiomyocytes. The slope conductances of cardiac TREK-1 and CHO/TREK-1 channels were 123 ± 7 and 113 ± 17 pS, respectively. The TREK-1 inhibitor L-3-n-butylphthalide (10 &mgr;M) reduced the currents in CHO/TREK-1 cells, normal cardiomyocytes, and hypertrophic cardiomyocytes by 48.5%, 54.3%, and 55.5%, respectively. The percentage of L-3-n-butylphthalide–inhibited outward whole-cell current in hypertrophic cardiomyocytes (23.7%) was larger than that in normal cardiomyocytes (14.2%). The percentage of chloroform-activated outward whole-cell current in hypertrophic cardiomyocytes (58.3%) was also larger than normal control (40.2%). Our results demonstrated that in hypertrophic rats, TREK-1 protein expression in endocardium was specifically increased and the ratio of TREK-1 channel current in cardiac outward currents was also enhanced. TREK-1 might balance potassium ion flow during hypertrophy and might be a potential drug target for heart protection.


European Journal of Pharmacology | 2012

Potassium 2-(1-hydroxypentyl)-benzoate attenuated hydrogen peroxide-induced apoptosis in neuroblastoma SK-N-SH cells.

Yanli Hu; Ying Peng; Yan Long; Shaofeng Xu; Nan Feng; Ling Wang; Xiaoliang Wang

Potassium 2-(1-hydroxypentyl)-benzoate (dl-PHPB) has been shown to have potent neuroprotective effects, such as reducing the infarct volume and improving neurobehavioral deficits in the transient focal cerebral ischemic rat model. The present study is to evaluate the neuroprotective effect of dl-PHPB on hydrogen peroxide (H(2)O(2))-induced apoptosis and the possible mechanism in the human neuroblastoma SK-N-SH cells. Our results showed that dl-PHPB significantly attenuated H(2)O(2)-induced cell death, and reduced neuronal apoptosis. Dl-PHPB partially reversed the decrease of B-cell CLL/lymphoma 2 (Bcl-2) protein level induced by H(2)O(2). Furthermore, dl-PHPB inhibited the elevation of pro-apoptotic Bcl-2-associated X protein (Bax) and caspase3, and alleviated the down-regulation of protein kinase C alpha (PKCα). The PKC inhibitor, Calphostin C significantly attenuated the protective effects of dl-PHPB. The findings suggest that dl-PHPB may protect neurons against H(2)O(2)-induced apoptosis by modulating apoptosis-related proteins, and PKC signaling pathway may be involved in the neuroprotection of dl-PHPB.


Biochemical and Biophysical Research Communications | 2008

Striatal 19S Rpt6 deficit is related to α-synuclein accumulation in MPTP-treated mice

Bin Liu; Qiaoqiao Shi; Shiping Ma; Nan Feng; Jiang Li; Ling Wang; Xiaoliang Wang

Striatal mitochondrial proteins were investigated using proteomics in the 1-methyl 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinsons disease. Four proteins, 19S proteasome ATPase Rpt6 (19S Rpt6), Lectin-related nature killer cell receptor LY 49S, Zinc finger A20 domain containing 1, and the sodium channel-associated protein 1 isoform 2, were significantly decreased while alpha-synuclein was increased in MPTP-treated mice. The altered levels of 19S Rpt6 and alpha-synuclein were further verified by Western blot. Experiments using small interfering RNA (siRNA) showed that alpha-synuclein was increased by 50% in cultured striatal neurons when 19S Rpt6 was knocked down. Taken together, our results imply that a deficiency in 19S Rpt6 may be partially related to the MPTP-induced increase in alpha-synuclein in the striatum.


Acta Pharmaceutica Sinica B | 2016

Parishin C's prevention of Aβ1–42-induced inhibition of long-term potentiation is related to NMDA receptors

Zhihui Liu; Weiping Wang; Nan Feng; Ling Wang; Jiangong Shi; Xiaoliang Wang

The rhizome of Gastrodia elata (GE), a herb medicine, has been used for treatment of neuronal disorders in Eastern Asia for hundreds of years. Parishin C is a major ingredient of GE. In this study, the i.c.v. injection of soluble Aβ1–42 oligomers model of LTP injury was used. We investigated the effects of parishin C on the improvement of LTP in soluble Aβ1–42 oligomer–injected rats and the underlying electrophysiological mechanisms. Parishin C (i.p. or i.c.v.) significantly ameliorated LTP impairment induced by i.c.v. injection of soluble Aβ1–42 oligomers. In cultured hippocampal neurons, soluble Aβ1–42 oligomers significantly inhibited NMDAR currents while not affecting AMPAR currents and voltage-dependent currents. Pretreatment with parishin C protected NMDA receptor currents from the damage induced by Aβ. In summary, parishin C improved LTP deficits induced by soluble Aβ1–42 oligomers. The protection by parishin C against Aβ-induced LTP damage might be related to NMDA receptors.


Acta Pharmacologica Sinica | 2018

Conversion and pharmacokinetics profiles of a novel pro-drug of 3- n -butylphthalide, potassium 2-(1-hydroxypentyl)-benzoate, in rats and dogs

Jiang Li; Shaofeng Xu; Ying Peng; Nan Feng; Ling Wang; Xiaoliang Wang

Potassium 2-(1-hydroxypentyl)-benzoate (dl-PHPB) is a novel pro-drug of 3-n-butylphthalide (dl-NBP) that is used to treat ischemic stroke. Currently, dl-PHPB is in phase II–III clinical trials in China. In this study, we investigated the conversion and pharmacokinetics profiles of dl-PHPB in vitro and in vivo. The conversion of dl-PHPB to dl-NBP was pH- and calcium-dependent, and paraoxonase was identified as a major enzyme for the conversion in rat plasma. The pharmacokinetics, tissue distribution and excretion of dl-PHPB were studied and compared with equal-molar doses of dl-NBP in rats and dogs. The in vivo studies showed that dl-PHPB could be quickly and completely converted to dl-NBP. The plasma concentration-time course of converted dl-NBP after intravenous dl-PHPB administration was nearly the same as that after equal-molar dl-NBP. The Cmax and AUC of dl-NBP after oral dl-PHPB administration in rats and dogs were higher by 60% and 170%, respectively, than those after oral dl-NBP administration. Analysis of the tissue distribution of dl-PHPB revealed that converted dl-NBP was primarily distributed in fat, the brain and the stomach. In the brain, the levels of dl-NBP were relatively higher after dl-PHPB treatment by orally than after treatment with equal-molar dl-NBP. Approximately 3%–4% of dl-NBP was excreted within 72 h after dosing with dl-PHPB or dl-NBP, but no dl-PHPB was detected in urine or feces excrements. Our results demonstrate that the conversion of dl-PHPB is fast after oral or intravenous administration. Furthermore, the bioavailability of dl-PHPB was obviously better than that of dl-NBP.


Journal of Alzheimer's Disease | 2017

Changes in Synaptic Plasticity and Glutamate Receptors in Type 2 Diabetic KK-Ay Mice

Huajing Yin; Weiping Wang; Wenwen Yu; Jiang Li; Nan Feng; Ling Wang; Xiaoliang Wang

In the present study, the progressive alteration of cognition and the mechanisms of reduction in long-term potentiation (LTP) in spontaneous obese KK-Ay type 2 diabetic mice were investigated. In the study, 3-, 5-, and 7-month-old KK-Ay mice were used. The results indicated that KK-Ay mice showed cognitive deficits in the Morris water maze test beginning at the age of 3 months. LTP was significantly impaired in KK-Ay mice during whole study period (3 to 7 months). The above deficits were reversible at an early stage (3 to 5 months old) by diet intervention. Moreover, we found the underlying mechanisms of LTP impairment in KK-Ay mice might be attributed to abnormal phosphorylation or expression of postsynaptic glutamate receptor subunits instead of alteration of basal synaptic transmission. The expression levels of NR1, NR2A, and NR2B subunits of N-methyl-d-aspartate receptors (NMDARs) were unchanged while the Tyr-dependent phosphorylation of both NR2A and NR2B subunits were significantly reduced in KK-Ay mice. The level of p-Src expression mediating this process was decreased, and the level of αCaMKII autophosphorylation was also reduced. Meanwhile, the GluR1 of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) was decreased, and GluR2 was significantly increased. These data suggest that deficits in synaptic plasticity in KK-Ay mice may arise from the abnormal phosphorylation of the NR2 subunits and the alteration of subunit composition of AMPARs. Diet intervention at an early stage of diabetes might alleviate the cognitive deficits and LTP reduction in KK-Ay mice.


Neuroscience Letters | 2018

Lig4-4 selectively inhibits TREK-1 and plays potent neuroprotective roles in vitro and in rat MCAO model

Weiping Wang; Dong-Mei Liu; Qiong Xiao; Jie Cai; Nan Feng; Shaofeng Xu; Ling Wang; Dali Yin; Xiaoliang Wang

Two-pore domain potassium channel TREK-1 was known to play an important role in neuroprotection, especially in acute cerebral ischemia. In the present study we found that 3-(nitromethyl) isobenzofuran-1(3H)-one (lig4-4) could robustly inhibit TREK-1 currents with an IC50 of 2.06 μM. However, the IC50 of lig4-4 for other ion channel subtypes such as Kv2.1, Kv1.5, Kv3.1, hERG and neuronal Na+ and Ca2+ channels were more than 30 μM, suggesting a specific inhibition of TREK-1 channel. MTT assay showed that lig4-4 significantly enhanced cell viability of cultured neurons under the condition of oxygen and glucose deprivation followed by reoxygenation (OGD/R). Annexin V/Propidium Iodide apoptosis assay also demonstrated that lig4-4 obviously reduced cell apoptosis in OGD/R-injured neurons. Western blotting results indicated that OGD/R-induced up-regulation of cleaved-caspase-3 expression and down-regulation of Bcl-2 could be notably reversed by lig4-4. The in vivo study showed that after oral administration of lig4-4 at 50 mg/kg, the infarct volume in middle cerebral artery occlusion (MCAO) rat model reduced from a vehicle control of 38.8% to 28%. Both in vitro and in vivo studies suggested that inhibition of TREK-1 with lig4-4 might produce a neuroprotective effect against cerebral ischemia. In conclusion, we demonstrated that lig4-4 selectively inhibited TREK-1 and protected brain from cerebral ischemic injury. The mechanisms might relate to block TREK-1 and inhibit neuronal apoptosis by modulating the expressions of Bcl-2 and cleaved-caspase-3.

Collaboration


Dive into the Nan Feng's collaboration.

Top Co-Authors

Avatar

Ling Wang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Xiaoliang Wang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Shaofeng Xu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jiang Li

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Shiping Ma

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Weiping Wang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Ying Peng

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jinghua Yang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Wanhong Zhao

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Yanli Hu

Peking Union Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge