Network


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

Hotspot


Dive into the research topics where Dan Ke is active.

Publication


Featured researches published by Dan Ke.


Glia | 2012

Upregulation of astrocytes protein phosphatase-2A stimulates astrocytes migration via inhibiting p38 MAPK in tg2576 mice.

Xiu-Ping Liu; Hong-Yun Zheng; Min Qu; Yao Zhang; Fu-Yuan Cao; Qun Wang; Dan Ke; Gong-Ping Liu; Jian-Zhi Wang

One of the earliest neuropathological changes in Alzheimer disease (AD) is the accumulation of astrocytes at sites of β‐amyloid (Aβ) deposits, but the cause of this cellular response is unclear. As the activity of protein phosphatase 2A (PP2A) is significantly decreased in the AD brains, we studied the role of PP2A in astrocytes migration. We observed unexpectedly that PP2A activity associated with glial fibrillary acidic protein, an astrocyte marker, was significantly upregulated in tg2576 mice, demonstrated by an increased enzyme activity, a decreased demethylation at leucine‐309 (DM‐PP2Ac), and a decreased phosphorylation at tyrosine‐307 of PP2A (pY307‐PP2Ac). Further studies by using in vitro wound‐healing model and transwell assay demonstrated that upregulation of PP2A pharmacologically and genetically could stimulate astrocytes migration. Activation of PP2A promotes actin organization and inhibits p38 mitogen‐activated protein kinases (p38 MAPK), while simultaneous activation of p38 MAPK partially abolishes the PP2A‐induced astrocytes migration. Our data suggest that activation of astrocytes PP2A in tg2567 mice may stimulate the migration of astrocytes to the amyloid plaques by p38 MAPK inhibition, implying that PP2A deficits observed in AD may cause Aβ accumulation via hindering the astrocytes migration.


Nature Communications | 2016

Opposite monosynaptic scaling of BLP–vCA1 inputs governs hopefulness- and helplessness-modulated spatial learning and memory

Ying Yang; Zhi-Hao Wang; Sen Jin; Di Gao; Nan Liu; Shanping Chen; Sinan Zhang; Qing Liu; Enjie Liu; Xin Wang; Xiao Liang; Pengfei Wei; Xiao-Guang Li; Yin Li; Chenyu Yue; Hong-Lian Li; Wang Y; Qun Wang; Dan Ke; Qingguo Xie; Fuqiang Xu; Liping Wang; Jian-Zhi Wang

Different emotional states lead to distinct behavioural consequences even when faced with the same challenging events. Emotions affect learning and memory capacities, but the underlying neurobiological mechanisms remain elusive. Here we establish models of learned helplessness (LHL) and learned hopefulness (LHF) by exposing animals to inescapable foot shocks or with anticipated avoidance trainings. The LHF animals show spatial memory potentiation with excitatory monosynaptic upscaling between posterior basolateral amygdale (BLP) and ventral hippocampal CA1 (vCA1), whereas the LHL show memory deficits with an attenuated BLP–vCA1 connection. Optogenetic disruption of BLP–vCA1 inputs abolishes the effects of LHF and impairs synaptic plasticity. By contrast, targeted BLP–vCA1 stimulation rescues the LHL-induced memory deficits and mimics the effects of LHF. BLP–vCA1 stimulation increases synaptic transmission and dendritic plasticity with the upregulation of CREB and intrasynaptic AMPA receptors in CA1. These findings indicate that opposite excitatory monosynaptic scaling of BLP–vCA1 controls LHF- and LHL-modulated spatial memory, revealing circuit-specific mechanisms linking emotions to memory.


EBioMedicine | 2016

Biomarkers for Early Diagnostic of Mild Cognitive Impairment in Type-2 Diabetes Patients: A Multicentre, Retrospective, Nested Case–Control Study

Zhi-Peng Xu; Su-Lian Yang; Shi Zhao; Cheng-Hong Zheng; Hong-Hua Li; Yao Zhang; Rong-Xi Huang; Mengzhu Li; Yuan Gao; Shujuan Zhang; Pei-Yan Zhan; Li-Fang Zhang; Lin Deng; Sheng Wei; Yanchao Liu; Jing-Wang Ye; Hu-Jun Ren; Na Li; Cai-Xia Kong; Xin Wang; Lin Fang; Qiuzhi Zhou; Hong-Wei Jiang; Jing-Rong Li; Qun Wang; Dan Ke; Gong-Ping Liu; Jian-Zhi Wang

Background Both type 2 diabetes mellitus (T2DM) and Alzheimers disease (AD) are common age-associated disorders and T2DM patients show an increased risk to suffer from AD, however, there is currently no marker to identify who in T2DM populations will develop AD. Since glycogen synthase kinase-3β (GSK-3β) activity, ApoE genotypes and olfactory function are involved in both T2DM and AD pathogenesis, we investigate whether alterations of these factors can identify cognitive impairment in T2DM patients. Methods The cognitive ability was evaluated using Minimum Mental State Examination (MMSE) and Clinical Dementia Rating (CDR), and the mild cognitive impairment (MCI) was diagnosed by Petersens criteria. GSK-3β activity in platelet, ApoE genotypes in leucocytes and the olfactory function were detected by Western/dot blotting, the amplification refractory mutation system (ARMS) PCR and the Connecticut Chemosensory Clinical Research Center (CCCRC) test, respectively. The odds ratio (OR) and 95% confidence intervals (95% CI) of the biomarkers for MCI diagnosis were calculated by logistic regression. The diagnostic capability of the biomarkers was evaluated by receiver operating characteristics (ROC) analyses. Findings We recruited 694 T2DM patients from Jan. 2012 to May. 2015 in 5 hospitals (Wuhan), and 646 of them met the inclusion criteria and were included in this study. 345 patients in 2 hospitals were assigned to the training set, and 301 patients in another 3 hospitals assigned to the validation set. Patients in each set were randomly divided into two groups: T2DM without MCI (termed T2DM-nMCI) or with MCI (termed T2DM-MCI). There were no significant differences for sex, T2DM years, hypertension, hyperlipidemia, coronary disease, complications, insulin treatment, HbA1c, ApoE ε2, ApoE ε3, tGSK3β and pS9GSK3β between the two groups. Compared with the T2DM-nMCI group, T2DM-MCI group showed lower MMSE score with older age, ApoE ε4 allele, higher olfactory score and higher rGSK-3β (ratio of total GSK-3β to Ser9-phosphorylated GSK-3β) in the training set and the validation set. The OR values of age, ApoE ε4 gene, olfactory score and rGSK-3β were 1.09, 2.09, 1.51, 10.08 in the training set, and 1.06, 2.67, 1.47, 7.19 in the validation set, respectively. The diagnostic accuracy of age, ApoE ε4 gene, olfactory score and rGSK-3β were 0.76, 0.72, 0.66, 0.79 in the training set, and 0.70, 0.68, 0.73, 0.79 in the validation set, respectively. These four combined biomarkers had the area under the curve (AUC) of 82% and 86%, diagnostic accuracy of 83% and 81% in the training set and the validation set, respectively. Interpretation Aging, activation of peripheral circulating GSK-3β, expression of ApoE ε4 and increase of olfactory score are diagnostic for the mild cognitive impairment in T2DM patients, and combination of these biomarkers can improve the diagnostic accuracy.


Neuroscience Bulletin | 2018

Tau-Induced Ca2+/Calmodulin-Dependent Protein Kinase-IV Activation Aggravates Nuclear Tau Hyperphosphorylation

Yu-Ping Wei; Jinwang Ye; Xiong Wang; Li-Ping Zhu; Qing-Hua Hu; Qun Wang; Dan Ke; Qing Tian; Jian-Zhi Wang

Hyperphosphorylated tau is the major protein component of neurofibrillary tangles in the brains of patients with Alzheimer’s disease (AD). However, the mechanism underlying tau hyperphosphorylation is not fully understood. Here, we demonstrated that exogenously expressed wild-type human tau40 was detectable in the phosphorylated form at multiple AD-associated sites in cytoplasmic and nuclear fractions from HEK293 cells. Among these sites, tau phosphorylated at Thr205 and Ser214 was almost exclusively found in the nuclear fraction at the conditions used in the present study. With the intracellular tau accumulation, the Ca2+ concentration was significantly increased in both cytoplasmic and nuclear fractions. Further studies using site-specific mutagenesis and pharmacological treatment demonstrated that phosphorylation of tau at Thr205 increased nuclear Ca2+ concentration with a simultaneous increase in the phosphorylation of Ca2+/calmodulin-dependent protein kinase IV (CaMKIV) at Ser196. On the other hand, phosphorylation of tau at Ser214 did not significantly change the nuclear Ca2+/CaMKIV signaling. Finally, expressing calmodulin-binding protein-4 that disrupts formation of the Ca2+/calmodulin complex abolished the okadaic acid-induced tau hyperphosphorylation in the nuclear fraction. We conclude that the intracellular accumulation of phosphorylated tau, as detected in the brains of AD patients, can trigger nuclear Ca2+/CaMKIV signaling, which in turn aggravates tau hyperphosphorylation. Our findings provide new insights for tauopathies: hyperphosphorylation of intracellular tau and an increased Ca2+ concentration may induce a self-perpetuating harmful loop to promote neurodegeneration.


Molecular Neurodegeneration | 2017

Downregulating ANP32A rescues synapse and memory loss via chromatin remodeling in Alzheimer model

Gao-Shang Chai; Qiong Feng; Zhi-Hao Wang; Yu Hu; Dong-Sheng Sun; Xiao-Guang Li; Dan Ke; Hong-Lian Li; Gong-Ping Liu; Jian-Zhi Wang

BackgroundThe impairment of histone acetylation is causally linked to the cognitive decline in Alzheimer’s disease (AD). In addition to histone acetyltransferases (HATs) and histone deacetylases (HDACs), inhibitor of acetyltransferases (INHAT) can also regulate histone acetylation. As a key component of INHAT, level of ANP32A is selectively upregulated in the brain of AD patients. Here we investigated whether downregulating ANP32A can rescue AD-like synapse and memory deficits.MethodsRFP-labeled lentiviral ANP32A-shRNA was infused stereotaxically into the hippocampal CA3 region of the human tau transgenic mice (termed htau). The spatial learning and memory were assessed by Morris water maze (MWM). The synaptic function was measured by electrophysiological recording and the spine density was detected by Golgi staining. RT-PCR and Western blotting were used to detect the mRNA and protein levels.ResultsElevation of ANP32 in htau transgenic mice was correlated with learning deficits, while the hippocampal infusion of lenti-siANP32A to downregulate ANP32A in 12 m-old htau mice could rescue memory loss. Further studies demonstrated that downregulating ANP32A restored synapse morphology and the function. In the brain of htau mice, the acetylated histone decreased while knockdown ANP32A unmasked histone for a robust acetylation with reduced INHAT complex formation. Downregulating of ANP32A also attenuated AD-like tau hyperphosphorylation. Finally, several AD-associated risk factors, including tau accumulation, β-amyloid and H2O2 exposure, increased ANP32A by activating CCAAT/enhancer binding protein-β (C/EBPβ).ConclusionWe conclude that downregulating ANP32A rescues synaptic plasticity and memory ability by reducing INHAT formation and unmasking histone for hyperacetylation. Our findings reveal novel mechanisms for AD memory loss and potential molecular markers for protection.


Proceedings of the National Academy of Sciences of the United States of America | 2018

BACE1 SUMOylation increases its stability and escalates the protease activity in Alzheimer’s disease

Jian Bao; Min Qin; Yacoubou Abdoul Razak Mahaman; Bin Zhang; Fang Huang; Kuan Zeng; Yiyuan Xia; Dan Ke; Qun Wang; Rong Liu; Jian-Zhi Wang; Keqiang Ye; Xiaochuan Wang

Significance BACE1 is a rate-limiting enzyme for amyloid beta polypeptide production, which plays a crucial role in Alzheimer’s disease (AD) pathogenesis. However, how this essential protease is posttranslationally regulated remains incompletely understood. In the current study, we show that K501 residue on BACE1, a ubiquitin modification site, is also competitively SUMOylated. We discovered that SUMOylation of BACE1 augments its stability and enzymatic activity, resulting in senile plaque formation and cognitive defect. Identification of the posttranslational modification on BACE1 provides insight into the molecular mechanism in AD. Amyloid beta (Aβ) is a major pathological marker in Alzheimer’s disease (AD), which is principally regulated by the rate-limiting β-secretase (i.e., BACE1) cleavage of amyloid precursor protein (APP). However, how BACE1 activity is posttranslationally regulated remains incompletely understood. Here, we show that BACE1 is predominantly SUMOylated at K501 residue, which escalates its protease activity and stability and subsequently increases Aβ production, leading to cognitive defect seen in the AD mouse model. Compared with a non-SUMOylated K501R mutant, injection of wild-type BACE1 significantly increases Aβ production and triggers cognitive dysfunction. Furthermore, overexpression of wild-type BACE1, but not non-SUMOylated K501R mutant, facilitates senile plaque formation and aggravates the cognitive deficit seen in the APP/PS1 AD mouse model. Together, our data strongly suggest that K501 SUMOylation on BACE1 plays a critical role in mediating its stability and enzymatic activity.


Neuropharmacology | 2017

Fluoxetine administration during adolescence attenuates cognitive and synaptic deficits in adult 3×TgAD mice

Dong-Sheng Sun; Lifeng Gao; Li Jin; Hao Wu; Qun Wang; You Zhou; Shuhao Fan; Xia Jiang; Dan Ke; Hao Lei; Jian-Zhi Wang; Gong-Ping Liu

&NA; Fluoxetine (FLX) has broad neurobiological functions and neuroprotective effects; however, the preventive effects of FLX on cognitive impairments in Alzheimers disease (AD) have not been reported. Here, we studied whether adolescent administration of fluoxetine can prevent memory deficits in AD transgenic mice that harbour PS1m146v, APPswe and TauP301L mutations (3 × TgAD). FLX was applied through peritoneal injection to the mice at postnatal day 35 (p35) for 15 consecutive days, and the effects of FLX were observed at 6‐month. We found that adolescent administration of FLX improved learning and memory abilities in 6‐month‐old 3 × TgAD mice. FLX exposure also increased the sizes of the hippocampal CA1, dentate gyrus (DG) and extensive cortex regions, with increased numbers of neurons and higher dendritic spine density. Meanwhile, the synaptic plasticity of neurons in the hippocampus was remodelled, and the expression levels of synaptic‐related proteins were increased along with activation of the cyclic AMP response element‐binding (CREB) protein/brain‐derived neurotrophic factor (BDNF) signalling pathway. Finally, we found that FLX effectively prevented the increase of beta‐amyloid (A&bgr;) levels. These data suggest that adolescent administration of the antidepressant drug FLX can efficiently preserve cognitive functions and improve pathologies in 3×Tg AD mice. HighlightsFLX exposure during adolescence prevented cognitive deficits of 6 m‐old 3 × TgAD mice.FLX exposure increased the sizes of hippocampal CA1 and dentate gyrus regions.FLX exposure remodelled the synaptic plasticity.


Frontiers in Aging Neuroscience | 2017

Knockdown of pp32 Increases Histone Acetylation and Ameliorates Cognitive Deficits

Qiong Feng; Gao-Shang Chai; Zhi-Hao Wang; Yu Hu; Dong-Sheng Sun; Xiao-Guang Li; Rong-Hong Ma; Yi-Rong Li; Dan Ke; Jian-Zhi Wang; Gong-Ping Liu

Aging is a cause of cognitive decline in the elderly and the major risk factor for Alzheimers disease, however, aging people are not all destined to develop into cognitive deficits, the molecular mechanisms underlying this difference in cognition of aging people are obscure. Epigenetic modifications, particularly histone acetylation in the nervous system, play a critical role in regulation of gene expression for learning and memory. An inhibitor of acetyltransferases (INHAT) is reported to suppress histone acetylation via a histone-masking mechanism, and pp32 is a key component of INHAT complex. In the present study, we divided ~18 m-old aged mice into the cognitive-normal and the cognitive-impaired group by Morris water maze, and found that pp32 level was significantly increased in the hippocampus of cognitive-impaired aged mice. The mRNA and protein levels of synaptic-associated proteins decreased with reduced dendrite complexity and histone acetylation. Knockdown of pp32 rescued cognitive decline in cognitive-impaired aged mice with restoration of synaptic-associated proteins, the increase of spine density and elevation of histone acetylation. Our study reveals a novel mechanism underlying the aging-associated cognitive disturbance, indicating that suppression of pp32 might represent a promising therapeutic approach for learning and memory impairments.


Frontiers in Molecular Neuroscience | 2018

CK2 Phosphorylating I2PP2A/SET Mediates Tau Pathology and Cognitive Impairment

Qing Zhang; Yiyuan Xia; Yongjun Wang; Yangping Shentu; Kuan Zeng; Yacoubou Abdoul Razak Mahaman; Fang Huang; Mengjuan Wu; Dan Ke; Qun Wang; Bin Zhang; Rong Liu; Jian-Zhi Wang; Keqiang Ye; Xiaochuan Wang

Casein kinase 2 (CK2) is highly activated in Alzheimer disease (AD) and is associated with neurofibrillary tangles formation. Phosphorylated SET, a potent PP2A inhibitor, mediates tau hyperphosphorylation in AD. However, whether CK2 phosphorylates SET and regulates tau pathological phosphorylation in AD remains unclear. Here, we show that CK2 phosphorylating SET at Ser9 induced tau hyperphosphorylation in AD. We found that either Aβ treatment or tau overexpression stimulated CK2 activation leading to SET Ser9 hyperphosphorylation in neurons and animal models, while inhibition of CK2 by TBB abolished this event. Overexpression of CK2 in mouse hippocampus via virus injection induced cognitive deficit associated with SET Ser9 hyperphosphorylation. Injection of SET Ser9 phosphorylation mimetic mutant induced tau pathology and behavior impairments. Conversely co-injection of non-phosphorylated SET S9A with CK2 abolished the CK2 overexpression-induced AD pathology and cognitive deficit. Together, our data demonstrate that CK2 phosphorylates SET at Ser9 leading to SET cytoplasmic translocation and inhibition of PP2A resulting in tau pathology and cognitive impairments.


bioRxiv | 2018

Tau accumulation activates STAT1 triggering memory deficits via suppressing NMDA receptor expression

Xiao-Guang Li; Xiaoyue Hong; Wang Y; Shujuan Zhang; Junfei Zhang; Xia-Chun Li; Yanchao Liu; Dongshen Sun; Qiong Feng; Jinwang Ye; Yuan Gao; Dan Ke; Qun Wang; Hong-Lian Li; Keqiang Ye; Gongping Liu; Jian-Zhi Wang

Intracellular tau accumulation forming neurofibrillary tangles is hallmark pathology of Alzheimers disease (AD), but how tau accumulation induces synapse impairment is elusive. By overexpressing human full-length wildtype tau (termed hTau) to mimic tau abnormality as seen in the brain of sporadic AD patients, we found that hTau accumulation activated JAK2 to phosphorylate STAT1 (Signal Transducer and Activator of Transcription 1) at Tyr701 leading to STAT1 dimerization, nuclear translocation and its activation. STAT1 activation suppressed expression of N-methyl-D-aspartate receptors (NMDARs) through direct binding to the specific GAS element of GluN1, GluN2A and GluN2B promoters, while knockdown STAT1 by AAV-Cre in STAT1flox/flox mice or expressing dominant negative Y701F-STAT1 efficiently rescued hTau-induced suppression of NMDARs expression with amelioration of synaptic functions and memory performance. These findings indicate that hTau accumulation impairs synaptic plasticity through JAK2/STAT1-induced suppression of NMDARs expression, revealing a novel mechanism for hTau-associated synapse and memory deficits.

Collaboration


Dive into the Dan Ke's collaboration.

Top Co-Authors

Avatar

Jian-Zhi Wang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Qun Wang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Gong-Ping Liu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Dong-Sheng Sun

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Xiao-Guang Li

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Rong Liu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Xiaochuan Wang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Zhi-Hao Wang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Bin Zhang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Fang Huang

Huazhong University of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge