Network


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

Hotspot


Dive into the research topics where Zhao-Qian Teng is active.

Publication


Featured researches published by Zhao-Qian Teng.


Stem Cells | 2010

MicroRNA miR-137 regulates neuronal maturation by targeting ubiquitin ligase Mind Bomb-1

Richard D. Smrt; Keith E. Szulwach; Rebecca L. Pfeiffer; Xuekun Li; Weixiang Guo; Manavendra Pathania; Zhao-Qian Teng; Yuping Luo; Junmin Peng; Angélique Bordey; Peng Jin; Xinyu Zhao

The maturation of young neurons is regulated by complex mechanisms and dysregulation of this process is frequently found in neurodevepmental disorders. MicroRNAs have been implicated in several steps of neuronal maturation including dendritic and axonal growth, spine development, and synaptogenesis. We demonstrate that one brain‐enriched microRNA, miR‐137, has a significant role in regulating neuronal maturation. Overexpression of miR‐137 inhibits dendritic morphogenesis, phenotypic maturation, and spine development both in brain and cultured primary neurons. On the other hand, a reduction in miR‐137 had opposite effects. We further show that miR‐137 targets the Mind bomb one (Mib1) protein through the conserved target site located in the 3′ untranslated region of Mib1 messenger RNA. Mib1 is an ubiquitin ligase known to be important for neurodevelopment. We show that exogenously expressed Mib1 could partially rescue the phenotypes associated with miR‐137 overexpression. These results demonstrate a novel miRNA‐mediated mechanism involving miR‐137 and Mib1 that function to regulate neuronal maturation and dendritic morphogenesis during development. STEM Cells 2010;28:1060–1070


PLOS ONE | 2012

miR-132 Enhances Dendritic Morphogenesis, Spine Density, Synaptic Integration, and Survival of Newborn Olfactory Bulb Neurons

Manavendra Pathania; Juan Torres-Reveron; Lily D. Yan; Tomoki Kimura; Tiffany V. Lin; Valerie Gordon; Zhao-Qian Teng; Xinyu Zhao; Tudor A. Fulga; David Van Vactor; Angélique Bordey

An array of signals regulating the early stages of postnatal subventricular zone (SVZ) neurogenesis has been identified, but much less is known regarding the molecules controlling late stages. Here, we investigated the function of the activity-dependent and morphogenic microRNA miR-132 on the synaptic integration and survival of olfactory bulb (OB) neurons born in the neonatal SVZ. In situ hybridization revealed that miR-132 expression occurs at the onset of synaptic integration in the OB. Using in vivo electroporation we found that sequestration of miR-132 using a sponge-based strategy led to a reduced dendritic complexity and spine density while overexpression had the opposite effects. These effects were mirrored with respective changes in the frequency of GABAergic and glutamatergic synaptic inputs reflecting altered synaptic integration. In addition, timely directed overexpression of miR-132 at the onset of synaptic integration using an inducible approach led to a significant increase in the survival of newborn neurons. These data suggest that miR-132 forms the basis of a structural plasticity program seen in SVZ-OB postnatal neurogenesis. miR-132 overexpression in transplanted neurons may thus hold promise for enhancing neuronal survival and improving the outcome of transplant therapies.


Neuron | 2011

RNA-binding Protein FXR2 Regulates Adult Hippocampal Neurogenesis by Reducing Noggin Expression

Weixiang Guo; Li Zhang; Devin M. Christopher; Zhao-Qian Teng; Sarah R. Fausett; Chang-Mei Liu; Olivia L. George; John Klingensmith; Peng Jin; Xinyu Zhao

In adult mammalian brains, neurogenesis persists in the subventricular zone of the lateral ventricles (SVZ) and the dentate gyrus (DG) of the hippocampus. Although evidence suggest that adult neurogenesis in these two regions is subjected to differential regulation, the underlying mechanism is unclear. Here, we show that the RNA-binding protein FXR2 specifically regulates DG neurogenesis by reducing the stability of Noggin mRNA. FXR2 deficiency leads to increased Noggin expression and subsequently reduced BMP signaling, which results in increased proliferation and altered fate specification of neural stem/progenitor cells in DG. In contrast, Noggin is not regulated by FXR2 in the SVZ, because Noggin expression is restricted to the ependymal cells of the lateral ventricles, where FXR2 is not expressed. Differential regulation of SVZ and DG stem cells by FXR2 may be a key component of the mechanism that governs the different neurogenic processes in these two adult germinal zones.


The Journal of Neuroscience | 2014

Synaptic and Cognitive Improvements by Inhibition of 2-AG Metabolism Are through Upregulation of MicroRNA-188-3p in a Mouse Model of Alzheimer's Disease

Jian Zhang; Mei Hu; Zhao-Qian Teng; Ya-Ping Tang; Chu Chen

Abnormal accumulation of β-amyloid (Aβ) is the major neuropathological hallmark of Alzheimers disease (AD). However, the mechanisms underlying aberrant Aβ formation in AD remain unclear. We showed previously that inhibition of monoacylglycerol lipase (MAGL), the primary enzyme that metabolizes the endocannabinoid 2-arachidonoylglycerol (2-AG) in the brain, robustly reduces Aβ by inhibiting β-site amyloid precursor protein cleaving enzyme 1 (BACE1), a key enzyme responsible for Aβ formation. However, the molecular mechanisms responsible for suppression of BACE1 by inhibition of 2-AG metabolism are largely unknown. We demonstrate here that expression of the noncoding small RNA miR-188-3p that targets BACE1 was significantly downregulated both in the brains of AD humans and APP transgenic (TG) mice, a mouse model of AD. The downregulated miR-188-3p expression was restored by MAGL inhibition. Overexpression of miR-188-3p in the hippocampus reduced BACE1, Aβ, and neuroinflammation and prevented deteriorations in hippocampal basal synaptic transmission, long-term potentiation, spatial learning, and memory in TG mice. 2-AG-induced suppression of BACE1 was prevented by miR-188-3p loss of function. Moreover, miR-188-3p expression was upregulated by 2-AG or peroxisome proliferator-activated receptor-γ (PPARγ) agonists and suppressed by PPARγ antagonism or NF-κB activation. Reducing Aβ and neuroinflammation by MAGL inhibition was occluded by PPARγ antagonism. In addition, BACE1 suppression by 2-AG and PPARγ activation was eliminated by knockdown of NF-κB. Our study provides a novel molecular mechanism underlying improved synaptic and cognitive function in TG mice by 2-AG signaling, which upregulates miR-188-3p expression through PPARγ and NF-κB signaling pathway, resulting in suppressions of BACE1 expression and Aβ formation.


PLOS ONE | 2013

An epigenetic feedback regulatory loop involving microRNA-195 and MBD1 governs neural stem cell differentiation.

Chang-Mei Liu; Zhao-Qian Teng; Andrea L. McQuate; Emily M. Jobe; Christa C. Christ; Sergei J. von Hoyningen-Huene; Marie D. Reyes; Eric D. Polich; Yina Xing; Yue Li; Weixiang Guo; Xinyu Zhao

Background Epigenetic mechanisms, including DNA methylation, histone modification, and microRNAs, play pivotal roles in stem cell biology. Methyl-CpG binding protein 1 (MBD1), an important epigenetic regulator of adult neurogenesis, controls the proliferation and differentiation of adult neural stem/progenitor cells (aNSCs). We recently demonstrated that MBD1 deficiency in aNSCs leads to altered expression of several noncoding microRNAs (miRNAs). Methodology/Principal Findings Here we show that one of these miRNAs, miR-195, and MBD1 form a negative feedback loop. While MBD1 directly represses the expression of miR-195 in aNSCs, high levels of miR-195 in turn repress the expression of MBD1. Both gain-of-function and loss-of-function investigations show that alterations of the MBD1–miR-195 feedback loop tip the balance between aNSC proliferation and differentiation. Conclusions/Significance Therefore the regulatory loop formed by MBD1 and miR-195 is an important component of the epigenetic network that controls aNSC fate.


Journal of Cerebral Blood Flow and Metabolism | 2015

Inhibition of Monoacylglycerol Lipase Prevents Chronic Traumatic Encephalopathy-like Neuropathology in a Mouse Model of Repetitive Mild Closed Head Injury

Jian Zhang; Zhao-Qian Teng; Yunping Song; Mei Hu; Chu Chen

Emerging evidence suggests that the risk of developing chronic traumatic encephalopathy (CTE), a progressive neurodegenerative disease, is significantly increased in military personnel and contact sports players who have been exposed to repetitive trauma brain injury (TBI). Unfortunately there are no effective medications currently available for prevention and treatment of CTE. Here we demonstrate that inhibition of monoacylglycerol lipase (MAGL), the key enzyme that metabolizes the endocannabinoid 2-arachidonoylglycerol (2-AG) in the brain, significantly reduced CTE-like neuropathologic changes in a mouse model of repetitive mild closed head injury (rmCHI). Inhibition of 2-AG metabolism promoted neurologic recovery following rmCHI and reduced proinflammatory cytokines, astroglial reactivity, expression of amyloid precursor protein and the enzymes that make Aβ, as well as formation of Aβ. Importantly, neurodegeneration, TDP-43 protein aggregation, and tau phosphorylation, which are the neuropathologic hallmarks of CTE, were significantly suppressed by MAGL inactivation. Furthermore, alterations in expression of glutamate receptor subunits and impairments in basal synaptic transmission, long-term synaptic plasticity, and spatial learning and memory were recovered by inhibition of 2-AG metabolism in animals exposed to rmCHI. Our results suggest that MAGL inhibition, which boosts 2-AG and reduces 2-AG metabolites prostaglandins in the brain, may lead to a new therapy for CTE.


Stem cell reports | 2017

MiR-203 Interplays with Polycomb Repressive Complexes to Regulate the Proliferation of Neural Stem/Progenitor Cells

Pei-Pei Liu; Gang-Bin Tang; Ya-Jie Xu; Yu-Qiang Zeng; Shuang-Feng Zhang; Hong-Zhen Du; Zhao-Qian Teng; Chang-Mei Liu

Summary The polycomb repressive complexes 1 (PRC1) and 2 (PRC2) are two distinct polycomb group (PcG) proteins that maintain the stable silencing of specific sets of genes through chromatin modifications. Although the PRC2 component EZH2 has been known as an epigenetic regulator in promoting the proliferation of neural stem/progenitor cells (NSPCs), the regulatory network that controls this process remains largely unknown. Here we show that miR-203 is repressed by EZH2 in both embryonic and adult NSPCs. MiR-203 negatively regulates the proliferation of NSPCs. One of PRC1 components, Bmi1, is a downstream target of miR-203 in NSPCs. Conditional knockout of Ezh2 results in decreased proliferation ability of both embryonic and adult NSPCs. Meanwhile, ectopic overexpression of BMI1 rescues the proliferation defects exhibited by miR-203 overexpression or EZH2 deficiency in NSPCs. Therefore, this study provides evidence for coordinated function of the EZH2-miR-203-BMI1 regulatory axis that regulates the proliferation of NSPCs.


Frontiers in Molecular Neuroscience | 2017

The Histone H3K27 Demethylase UTX Regulates Synaptic Plasticity and Cognitive Behaviors in Mice

Gang Bin Tang; Yu Qiang Zeng; Pei Pei Liu; Ting Wei Mi; Shuang Feng Zhang; Shang Kun Dai; Qing Yuan Tang; Lin Yang; Ya Jie Xu; Hai Liang Yan; Hong Zhen Du; Zhao-Qian Teng; Feng Quan Zhou; Chang-Mei Liu

Histone demethylase UTX mediates removal of repressive trimethylation of histone H3 lysine 27 (H3K27me3) to establish a mechanistic switch to activate large sets of genes. Mutation of Utx has recently been shown to be associated with Kabuki syndrome, a rare congenital anomaly syndrome with dementia. However, its biological function in the brain is largely unknown. Here, we observe that deletion of Utx results in increased anxiety-like behaviors and impaired spatial learning and memory in mice. Loss of Utx in the hippocampus leads to reduced long-term potentiation and amplitude of miniature excitatory postsynaptic current, aberrant dendrite development and defective synapse formation. Transcriptional profiling reveals that Utx regulates a subset of genes that are involved in the regulation of dendritic morphology, synaptic transmission, and cognition. Specifically, Utx deletion disrupts expression of neurotransmitter 5-hydroxytryptamine receptor 5B (Htr5b). Restoration of Htr5b expression in newborn hippocampal neurons rescues the defects of neuronal morphology by Utx ablation. Therefore, we provide evidence that Utx plays a critical role in modulating synaptic transmission and cognitive behaviors. Utx cKO mouse models like ours provide a valuable means to study the underlying mechanisms of the etiology of Kabuki syndrome.


The Neuroscientist | 2018

Polycomb Repressive Complex 2: Emerging Roles in the Central Nervous System:

Pei-Pei Liu; Ya-Jie Xu; Zhao-Qian Teng; Chang-Mei Liu

The polycomb repressive complex 2 (PRC2) is responsible for catalyzing both di- and trimethylation of histone H3 at lysine 27 (H3K27me2/3). The subunits of PRC2 are widely expressed in the central nervous system (CNS). PRC2 as well as H3K27me2/3, play distinct roles in neuronal identity, proliferation and differentiation of neural stem/progenitor cells, neuronal morphology, and gliogenesis. Mutations or dysregulations of PRC2 subunits often cause neurological diseases. Therefore, PRC2 might represent a common target of different pathological processes that drive neurodegenerative diseases. A better understanding of the intricate and complex regulatory networks mediated by PRC2 in CNS will help to develop new therapeutic approaches and to generate specific brain cell types for treating neurological diseases.


Stem cell reports | 2018

A Chemical Recipe for Generation of Clinical-Grade Striatal Neurons from hESCs

Menghua Wu; Da Zhang; Chunying Bi; Tingwei Mi; Wenliang Zhu; Longkuo Xia; Zhao-Qian Teng; Baoyang Hu; Yihui Wu

Summary Differentiation of human pluripotent stem cells (hPSCs) into striatal medium spiny neurons (MSNs) promises a cell-based therapy for Huntingtons disease. However, clinical-grade MSNs remain unavailable. Here, we developed a chemical recipe named XLSBA to generate clinical-grade MSNs from embryonic stem cells (ESCs). We introduced the γ-secretase inhibitor DAPT into the recipe to accelerate neural differentiation, and replaced protein components with small molecules. Using this optimized protocol we could efficiently direct regular human ESCs (hESCs) as well as clinical-grade hESCs to lateral ganglionic eminence (LGE)-like progenitors and striatal MSNs within less than half of the time than previous protocols (within 14 days and 21 days, respectively). These striatal cells expressed appropriate MSN markers and electrophysiologically acted like authentic MSNs. Upon transplantation into brains of neonatal mice or mouse model of Huntingtons disease, they exhibited sufficient safety and reasonable efficacy. Therefore, this quick and highly efficient derivation of MSNs offers unprecedented access to clinical application.

Collaboration


Dive into the Zhao-Qian Teng's collaboration.

Top Co-Authors

Avatar

Chang-Mei Liu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xinyu Zhao

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Weixiang Guo

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Pei-Pei Liu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Ya-Jie Xu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gang-Bin Tang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Hong-Zhen Du

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Shuang-Feng Zhang

University of Science and Technology of China

View shared research outputs
Researchain Logo
Decentralizing Knowledge